(ALPHA-SUBSTITUTED CYCLOALKYLAMINO AND HETEROCYCLYLAMINO) PYRIMIDINYL AND 1,3,5-TRIAZINYL BENZIMIDAZOLES, PHARMACEUTICAL COMPOSITIONS THEREOF, AND THEIR USE IN TREATING PROLIFERATIVE DISEASES

- MEI PHARMA, INC.

Provided herein are (alpha-substituted cycloalkylamino or heterocyclylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, e.g., a compound of Formula I, and their pharmaceutical compositions, preparation, and use as agents or drugs for treating proliferative diseases.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of the priority of U.S. Provisional Application Nos. 61/468,506, filed Mar. 28, 2011; and 61/530,839, filed Sep. 2, 2011; the disclosure of each of which is incorporated herein by reference in its entirety.

FIELD

Provided herein are (alpha-substituted cycloalkylamino or heterocyclylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, and their pharmaceutical compositions, preparation, and use as agents or drugs for treating proliferative diseases.

BACKGROUND

Phosphoinositide-3-kinases (PI3Ks) are a group of lipid kinases, which phosphorylate the 3-hydroxyl of phosphoinositides. They are classified into at least three classes (classes I, II, and III) and play an important role in cellular signaling (Stephens et al., Curr. Opin. Pharmacol. 2005, 5, 357). Class I enzymes are further classified into classes Ia and Ib based on their mechanism of activation. Class Ia PI3Ks are heterodimeric structures consisting of a catalytic subunit (p110α, p110β, or p110δ in complex with a regulatory p85 subunit, while class-Ib PI3K (p110γ) is structurally similar but lacks the p85 regulatory subunit, and instead is activated by βγ subunits of heterotrimeric G-proteins (Walker et al., Mol. Cell. 2000, 6, 909).

PI3Ks play a variety of roles in normal tissue physiology (Foukas & Shepherd, Biochem. Soc. Trans. 2004, 32, 330; Shepherd, Acta Physiol. Scand. 2005, 183, 3), with p110α having a specific role in cancer growth, p110β in thrombus formation mediated by integrin αIIβ3 (Jackson et al., Nat. Med. 2005, 11, 507), and p110γ in inflammation, rheumatoid arthritis, and other chronic inflammation states (Barber et al., Nat. Med. 2005, 11, 933; Camps et al., Nat. Med. 2005, 11, 936; Rommel et al., Nat. Rev. 2007, 7, 191; and Ito, et al., J. Pharm. Exp. Therap. 2007, 321, 1). Therefore, there is a need for PI3K inhibitors for treating cancer and/or inflammatory diseases.

SUMMARY OF THE DISCLOSURE

Provided herein is a compound of Formula I:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:

X, Y, and Z are each independently N or CRX, with the proviso that at least two of X, Y, and Z are nitrogen atoms; where RX is hydrogen or C1-6 alkyl;

R1 and R2 are each independently (a) hydrogen, cyano, halo, or nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, —C(NR1a)NR1bR1c, —OR1a, —OC(O)R1a, —OC(O)OR1a, —OC(O)NR1bR1c, —OC(═NR1a)NR1bR1c, —OS(O)R1a, —OS(O)2R1a, —OS(O)NR1bR1c, —OS(O)2NR1bR1c, —NR1bR1c, —NR1aC(O)R1d, —NR1aC(O)OR1d, —NR1aC(O)NR1bR1c, —NR1aC(═NR1d)NR1bR1c, —NR1aS(O)R1d, —NR1aS(O)2R1d, —NR1aS(O)NR1bR1c, —NR1aS(O)2NR1bR1c, —SR1a, —S(O)R1a, —S(O)2R1a, —S(O)NR1bR1c, or —S(O)2NR1bR1c; wherein each R1a, R1b, R1c, and R1d is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) R1b and R1c together with the N atom to which they are attached form heterocyclyl;

R3 and R4 are each independently hydrogen or C1-6 alkyl; or R3 and R4 are linked together to form a bond, C1-6 alkylene, C1-6 heteroalkylene, C2-6 alkenylene, or C2-6 heteroalkenylene;

R5a and R5b together with the carbon atom to which they are attached form C3-10 cycloalkyl or heterocyclyl;

R5c is C6-14 aryl, heteroaryl, C7-15 aralkyl, or heteroaryl-C1-C6 alkyl; and

R6 is hydrogen, C1-6 alkyl, —S—C1-6 alkyl, —S(O)—C1-6 alkyl, or —SO2—C1-6 alkyl;

wherein each alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaryl-alkyl, and heterocyclyl in R1, R2, R3, R4, R6, RX, R1a, R1b, R1c, R1d, R5a, R5b, and R5c is optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q, wherein each substituent Q is independently selected from (a) oxo, cyano, halo, and nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; and (c) —C(O)Ra, —C(O)ORa, —C(O)NRbRc, —C(NRa)NR1bR1c, —ORa, —OC(O)Ra, —OC(O)ORa, —OC(O)NR1bR1c, —OC(═NRa)NRbRc, —OS(O)Ra, —OS(O)2R1a, —OS(O)NRbRc, —OS(O)2NRbRc, —NR1bR1c, —NRaC(O)Rd, —NRaC(O)ORd, —NRaC(O)NRbRc, —NRaC(═NRd)NRbRc, —NRaS(O)Rd, —NRaS(O)2Rd, —NRaS(O)NR1bR1c, —NRaS(O)2NR1bR1c, —SRa, —S(O)Ra, —S(O)2R1a, —S(O)NR1bR1c, and —S(O)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa;

wherein each Qa is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) —C(O)Re, —C(O)ORe, —C(O)NRfRg, —C(NRe)NRfRg, —ORe, —OC(O)Re, —OC(O)ORe, —OC(O)NRfRg, —OC(═NRe)NRfRg, —OS(O)Re, —OS(O)2Re, —OS(O)NRfRg, —OS(O)2NRfRg, —NRfRg, —NReC(O)Rh, —NReC(O)ORh, —NReC(O)NRfRg, —NReC(═NRh)NRfRg, —NReS(O)Rh, —NReS(O)2Rh, —NReS(O)NRfRg, —NReS(O)2NRfRg, —SRe, —S(O)Re, —S(O)2Re, —S(O)NRfRg, and —S(O)2NRfRg; wherein each Re, Rf, Rg, and Rh is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and Rg together with the N atom to which they are attached form heterocyclyl.

Also provided herein is a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:

X, Y, and Z are each independently N or CRX, with the proviso that at least two of X, Y, and Z are nitrogen atoms; where RX is hydrogen or C1-6 alkyl;

R1 and R2 are each independently (a) hydrogen, cyano, halo, or nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, —C(NR1a)NR1bR1c, —OR1a, —OC(O)R1a, —OC(O)OR1a, —OC(O)NR1bR1c, —OC(═NR1a)NR1bR1c, —OS(O)R1a, —OS(O)2R1a, —OS(O)NR1bR1c, —OS(O)2NR1bR1c, —NR1bR1c, —NR1aC(O)R1d, —NR1aC(O)ORd, —NR1aC(O)NR1bR1c, —NR1aC(═NR1d)NR1bR1c, —NR1aS(O)R1d, —NR1aS(O)2R1d, —NR1S(O)NR1bR1c, —NR1aS(O)2NR1bR1c, —SR1a, —S(O)R1a, —S(O)2R1a, —S(O)NR1bR1c, or —S(O)2NR1bR1c; wherein each R1a, R1b, R1c, and R1d is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) R1b and R1c together with the N atom to which they are attached form heterocyclyl;

R3 and R4 are each independently hydrogen or C1-6 alkyl; or R3 and R4 are linked together to form a bond, C1-6 alkylene, C1-6 heteroalkylene, C2-6 alkenylene, or C2-6 heteroalkenylene;

R5a and R5b together with the carbon atom to which they are attached form C3-10 cycloalkyl or heterocyclyl;

R5c is C6-14 aryl or heteroaryl; and

R6 is hydrogen, C1-6 alkyl, —S—C1-6 alkyl, —S(O)—C1-6 alkyl, or —SO2—C1-6 alkyl;

wherein each alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl in R1, R2, R3, R4, R6, RX, R1a, R1b, R1c, R1d, R5a, R5b, and R5c is optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q, wherein each substituent Q is independently selected from (a) oxo, cyano, halo, and nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; and (c) —C(O)Ra, —C(O)ORa, —C(O)NR1bR1c, —C(NRa)NR1bR1c, —ORa, —OC(O)Ra, —OC(O)ORa, —OC(O)NRbRc, —OC(═NRa)NRbRc, —OS(O)Ra, —OS(O)2R1a, —OS(O)NRbRc, —OS(O)2NRbRc, —NRbRc, —NRaC(O)Rd, —NRaC(O)ORd, —NRaC(O)NRbRc, —NRaC(═NRd)NRbRc, —NRaS(O)Rd, —NRaS(O)2Rd, —NRaS(O)NRbRc, —NRaS(O)2NRbRc, —SRa, —S(O)Ra, —S(O)2R1a, —S(O)NRbRc, and —S(O)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q;

wherein each Qa is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) —C(O)Re, —C(O)ORe, —C(O)NRfRg, —C(NRe)NRfRg, —ORe, —OC(O)Re, —OC(O)ORe, —OC(O)NRfRg, —OC(═NRe)NRfRg, —OS(O)Re, —OS(O)2Re, —OS(O)NRfRg, —OS(O)2NRfRg, —NRfRg, —NReC(O)Rh, —NReC(O)ORh, —NReC(O)NRfRg, —NReC(═NRh)NRfRg, —NReS(O)Rh, —NReS(O)2Rh, —NReS(O)NRfRg, —NReS(O)2NRfRg, —SRe, —S(O)Re, —S(O)2Re, —S(O)NRfRg, and —S(O)2NRfRg; wherein each Re, Rf, Rg, and Rh is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and Rg together with the N atom to which they are attached form heterocyclyl.

Additionally, provided herein are pharmaceutical compositions comprising a compound disclosed herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients.

Furthermore, provided herein is a method for treating, preventing, or ameliorating one or more symptoms of a PI3K-mediated disorder, disease, or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

Provided herein is a method for treating, preventing, or ameliorating one or more symptoms of a PI3KS-mediated disorder, disease, or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

Provided herein is a method for treating, preventing, or ameliorating one or more symptoms of a proliferative disease in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

Provided herein is a method for modulating PI3K activity, comprising contacting a PI3K with an effective amount of a compound disclosed herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

Provided herein is a method for modulating PI3Kδ activity, comprising contacting PI3Kδ with an effective amount of a compound disclosed herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

Provided herein is a method for selectively modulating PI3Kδ activity, comprising contacting PI3Kδ with an effective amount of a compound disclosed herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

DETAILED DESCRIPTION

To facilitate understanding of the disclosure set forth herein, a number of terms are defined below.

Generally, the nomenclature used herein and the laboratory procedures in organic chemistry, medicinal chemistry, and pharmacology described herein are those well known and commonly employed in the art. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs.

The term “subject” refers to an animal, including, but not limited to, a primate (e.g., human), cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms “subject” and “patient” are used interchangeably herein in reference, for example, to a mammalian subject, such as a human subject, in one embodiment, a human.

The terms “treat,” “treating,” and “treatment” are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or alleviating or eradicating the cause(s) of the disorder, disease, or condition itself.

The terms “prevent,” “preventing,” and “prevention” are meant to include a method of delaying and/or precluding the onset of a disorder, disease, or condition, and/or its attendant symptoms; barring a subject from acquiring a disorder, disease, or condition; or reducing a subject's risk of acquiring a disorder, disease, or condition.

The term “therapeutically effective amount” are meant to include the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disorder, disease, or condition being treated. The term “therapeutically effective amount” also refers to the amount of a compound that is sufficient to elicit the biological or medical response of a biological molecule (e.g., a protein, enzyme, RNA, or DNA), cell, tissue, system, animal, or human, which is being sought by a researcher, veterinarian, medical doctor, or clinician.

The term “pharmaceutically acceptable carrier,” “pharmaceutically acceptable excipient,” “physiologically acceptable carrier,” or “physiologically acceptable excipient” refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or encapsulating material. In one embodiment, each component is “pharmaceutically acceptable” in the sense of being compatible with other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, Remington: The Science and Practice of Pharmacy, 21st Edition, Lippincott Williams & Wilkins: Philadelphia, Pa., 2005; Handbook of Pharmaceutical Excipients, 5th Edition, Rowe et al., Eds., The Pharmaceutical Press and the American Pharmaceutical Association: 2005; and Handbook of Pharmaceutical Additives, 3rd Edition, Ash and Ash Eds., Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, 2nd Edition, Gibson Ed., CRC Press LLC: Boca Raton, Fla., 2009.

The term “about” or “approximately” means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term “about” or “approximately” means within 1, 2, 3, or 4 standard deviations. In certain embodiments, the term “about” or “approximately” means within 50%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.05% of a given value or range.

The terms “active ingredient” and “active substance” refer to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients, to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder, disease, or condition. As used herein, “active ingredient” and “active substance” may be an optically active isomer of a compound described herein.

The terms “drug,” “therapeutic agent,” and “chemotherapeutic agent” refer to a compound, or a pharmaceutical composition thereof, which is administered to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder, disease, or condition.

The term “naturally occurring” or “native” when used in connection with biological materials such as nucleic acid molecules, polypeptides, host cells, and the like, refers to materials which are found in nature and are not manipulated by man. Similarly, “non-naturally occurring” or “non-native” refers to a material that is not found in nature or that has been structurally modified or synthesized by man.

The term “PI3K” refers to a phosphoinositide 3-kinase or variant thereof, which is capable of phosphorylating the inositol ring of PI in the D-3 position. The term “PI3K variant” is intended to include proteins substantially homologous to a native PI3K, i.e., proteins having one or more naturally or non-naturally occurring amino acid deletions, insertions, or substitutions (e.g., PI3K derivatives, homologs, and fragments), as compared to the amino acid sequence of a native PI3K. The amino acid sequence of a PI3K variant is at least about 80% identical, at least about 90% identical, or at least about 95% identical to a native PI3K. Examples of PI3K include, but are not limited to, p110α, p110β, p110δ, p110γ, PI3K-C2α, PI3K-C2β, PI3K-C2γ, Vps34, mTOR, ATM, ATR, and DNA-PK. See, Fry, Biochem. Biophys. Acta 1994, 1226, 237-268; Vanhaesebroeck and Waterfield, Exp. Cell. Res. 1999, 253, 239-254; and Fry, Breast Cancer Res. 2001, 3, 304-312. PI3Ks are classified into at least four classes. Class I includes p110α, p110β, p110δ, and p110γ. Class II includes PI3K-C2α, PI3K-C2β, and PI3K-C2γ. Class III includes Vps34. Class IV includes mTOR, ATM, ATR, and DNA-PK. In certain embodiments, the PI3K is a Class I kinase. In certain embodiments, the PI3K is p110α, p110β, p110δ, or p110γ. In certain embodiments, the PI3K is a variant of a Class I kinase. In certain embodiments, the PI3K is a p110α mutant. Examples of p110α mutants include, but are not limited to, R38H, G106V, K111N, K227E, N345K, C420R, P539R, E542K, E545A, E545G, E545K, Q546K, Q546P, E453Q, H710P, I800L, T1025S, M1043I, M1043V, H1047L, H1047R, and H1047Y (Ikenoue et al., Cancer Res. 2005, 65, 4562-4567; Gymnopoulos et al., Proc. Natl. Acad. Sci., 2007, 104, 5569-5574). In certain embodiments, the PI3K is a Class II kinase. In certain embodiments, the PI3K is PI3K-C2α, PI3K-C2β, or PI3K-C2γ. In certain embodiments, the PI3K is a Class III kinase. In certain embodiments, the PI3K is Vps34. In certain embodiments, the PI3K is a Class IV kinase. In certain embodiments, the PI3K is mTOR, ATM, ATR, or DNA-PK.

The terms “PI3K-mediated disorder, disease, or condition” and “a disorder, disease, or condition mediated by PI3K” refer to a disorder, disease, or condition characterized by abnormal or dysregulated, e.g., less than or greater than normal, PI3K activity. Abnormal PI3K functional activity might arise as the result of PI3K overexpression in cells, expression of PI3K in cells which normally do not express PI3K, or dysregulation due to constitutive activation, caused, for example, by a mutation in PI3K. A PI3K-mediated disorder, disease, or condition may be completely or partially mediated by abnormal PI3K activity. In particular, PI3K-mediated disorder, disease, or condition is one in which modulation of a PI3K activity results in some effect on the underlying disorder, disease, or condition, e.g., a PI3K inhibitor results in some improvement in at least some of patients being treated.

The terms “p110δ-mediated disorder, disease, or condition,” “a disorder, disease, or condition mediated by p110δ,” “PI3KS-mediated disorder, disease, or condition,” and “a disorder, disease, or condition mediated by PI3KS” refer to a disorder, disease, or condition characterized by abnormal or dysregulated, e.g., less than or greater than normal, p110δ activity. Abnormal p110δ functional activity might arise as the result of p110δ overexpression in cells, expression of p110 in cells which normally do not express p110δ, or dysregulation due to constitutive activation, caused, for example, by a mutation in p110δ. A p110δ-mediated disorder, disease, or condition may be completely or partially mediated by abnormal p110δ activity. In particular, p110δ-mediated disorder, disease, or condition is one in which modulation of a p110δ activity results in some effect on the underlying disorder, disease, or condition, e.g., a p110δ inhibitor results in some improvement in at least some of patients being treated.

The term “alkyl” refers to a linear or branched saturated monovalent hydrocarbon radical, wherein the alkylene may optionally be substituted with one or more substituents Q as described herein. The term “alkyl” also encompasses both linear and branched alkyl, unless otherwise specified. In certain embodiments, the alkyl is a linear saturated monovalent hydrocarbon radical that has 1 to 20 (C1-20), 1 to 15 (C1-C5), 1 to 10 (C1-10), or 1 to 6 (C1-6) carbon atoms, or branched saturated monovalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. As used herein, linear C1-6 and branched C3-6 alkyl groups are also referred as “lower alkyl.” Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl (including all isomeric forms), n-propyl, isopropyl, butyl (including all isomeric forms), n-butyl, isobutyl, sec-butyl, t-butyl, pentyl (including all isomeric forms), and hexyl (including all isomeric forms). For example, C1-6 alkyl refers to a linear saturated monovalent hydrocarbon radical of 1 to 6 carbon atoms or a branched saturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.

The term “alkylene” refers to a linear or branched saturated divalent hydrocarbon radical, wherein the alkylene may optionally be substituted with one or more substituents Q as described herein. The term “alkylene” encompasses both linear and branched alkylene, unless otherwise specified. In certain embodiments, the alkylene is a linear saturated divalent hydrocarbon radical that has 1 to 20 (C1-20), 1 to 15 (C1-C15), 1 to 10 (C1-10), or 1 to 6 (C1-6) carbon atoms, or branched saturated divalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. As used herein, linear C1-6 and branched C3-6 alkylene groups are also referred as “lower alkylene.” Examples of alkylene groups include, but are not limited to, methylene, ethylene, propylene (including all isomeric forms), n-propylene, isopropylene, butylene (including all isomeric forms), n-butylene, isobutylene, t-butylene, pentylene (including all isomeric forms), and hexylene (including all isomeric forms). For example, C1-6 alkylene refers to a linear saturated divalent hydrocarbon radical of 1 to 6 carbon atoms or a branched saturated divalent hydrocarbon radical of 3 to 6 carbon atoms.

The term “heteroalkylene” refers to a linear or branched saturated divalent hydrocarbon radical that contains one or more heteroatoms each independently selected from O, S, and N in the hydrocarbon chain. For example, C1-6 heteroalkylene refers to a linear saturated divalent hydrocarbon radical of 1 to 6 carbon atoms or a branched saturated divalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the heteroalkylene is a linear saturated divalent hydrocarbon radical that has 1 to 20 (C1-20), 1 to 15 (C1-C15), 1 to 10 (C1-10), or 1 to 6 (C1-6) carbon atoms, or branched saturated divalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. As used herein, linear C1-6 and branched C3-6 heteroalkylene groups are also referred as “lower heteroalkylene.” Examples of heteroalkylene groups include, but are not limited to, —CH2O—, —CH2OCH2—, —CH2CH2O—, —CH2NH—, —CH2NHCH2—, —CH2CH2NH—, —CH2S—, —CH2SCH2—, and —CH2CH2S—. In certain embodiments, heteroalkylene may also be optionally substituted with one or more substituents Q as described herein.

The term “alkenyl” refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one, two, three, four, or five, in another embodiment, one, carbon-carbon double bond(s). The alkenyl may be optionally substituted with one or more substituents Q as described herein. The term “alkenyl” also embraces radicals having “cis” and “trans” configurations, or alternatively, “Z” and “E” configurations, as appreciated by those of ordinary skill in the art. As used herein, the term “alkenyl” encompasses both linear and branched alkenyl, unless otherwise specified. For example, C2-6 alkenyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkenyl is a linear monovalent hydrocarbon radical of 2 to 20 (C2-20), 2 to 15 (C2-15), 2 to 10 (C2-10), or 2 to 6 (C2-6) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. Examples of alkenyl groups include, but are not limited to, ethenyl, propen-1-yl, propen-2-yl, allyl, butenyl, and 4-methylbutenyl.

The term “alkenylene” refers to a linear or branched divalent hydrocarbon radical, which contains one or more, in one embodiment, one, two, three, four, or five, in another embodiment, one, carbon-carbon double bond(s). The alkenylene may be optionally substituted with one or more substituents Q as described herein. Similarly, the term “alkenylene” also embraces radicals having “cis” and “trans” configurations, or alternatively, “E” and “Z” configurations. As used herein, the term “alkenylene” encompasses both linear and branched alkenylene, unless otherwise specified. For example, C2-6 alkenylene refers to a linear unsaturated divalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated divalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkenylene is a linear divalent hydrocarbon radical of 2 to 20 (C2-20), 2 to 15 (C2-15), 2 to 10 (C2-10), or 2 to 6 (C2-6) carbon atoms, or a branched divalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. Examples of alkenylene groups include, but are not limited to, ethenylene, allylene, propenylene, butenylene, and 4-methylbutenylene.

The term “heteroalkenylene” refers to a linear or branched divalent hydrocarbon radical, which contains one or more, in one embodiment, one, two, three, four, or five, in another embodiment, one, carbon-carbon double bond(s), and which contains one or more heteroatoms each independently selected from O, S, and N in the hydrocarbon chain. The heteroalkenylene may be optionally substituted with one or more substituents Q as described herein. The term “heteroalkenylene” embraces radicals having a “cis” or “trans” configuration or a mixture thereof, or alternatively, a “Z” or “E” configuration or a mixture thereof, as appreciated by those of ordinary skill in the art. For example, C2-6 heteroalkenylene refers to a linear unsaturated divalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated divalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the heteroalkenylene is a linear divalent hydrocarbon radical of 2 to 20 (C2-20), 2 to 15 (C2-15), 2 to 10 (C2-10), or 2 to 6 (C2-6) carbon atoms, or a branched divalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. Examples of heteroalkenylene groups include, but are not limited to, —CH═CHO—, —CH═CHOCH2—, —CH═CHCH2O—, —CH═CHS—, —CH═CHSCH2—, —CH═CHCH2S—, or —CH═CHCH2NH—.

The term “alkynyl” refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one, two, three, four, or five, in another embodiment, one, carbon-carbon triple bond(s). The alkynyl may be optionally substituted with one or more substituents Q as described herein. The term “alkynyl” also encompasses both linear and branched alkynyl, unless otherwise specified. In certain embodiments, the alkynyl is a linear monovalent hydrocarbon radical of 2 to 20 (C2-20), 2 to 15 (C2-15), 2 to 10 (C2-10), or 2 to 6 (C2-6) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl (—C≡CH) and propargyl (—CH2C≡CH). For example, C2-6 alkynyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.

The term “cycloalkyl” refers to a cyclic saturated bridged and/or non-bridged monovalent hydrocarbon radical, which may be optionally substituted with one or more substituents Q as described herein. In certain embodiments, the cycloalkyl has from 3 to 20 (C3-20), from 3 to 15 (C3-15), from 3 to 10 (C3-10), or from 3 to 7 (C3-7) carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, decalinyl, and adamantyl.

The term “cycloalkenyl” refers to a cyclic unsaturated, nonaromatic bridged and/or non-bridged monovalent hydrocarbon radical, which may be optionally substituted with one or more substituents Q as described herein. In certain embodiments, the cycloalkenyl has from 3 to 20 (C3-20), from 3 to 15 (C3-15), from 3 to 10 (C3-10), or from 3 to 7 (C3-7) carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclobutenyl, cyclopentenyl, cyclohexenyl, or cycloheptenyl,

The term “aryl” refers to a monocyclic aromatic group and/or multicyclic monovalent aromatic group that contain at least one aromatic hydrocarbon ring. In certain embodiments, the aryl has from 6 to 20 (C6-20), from 6 to 15 (C6-15), or from 6 to 10 (C6-10) ring atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, pyrenyl, biphenyl, and terphenyl. Aryl also refers to bicyclic or tricyclic carbon rings, where one of the rings is aromatic and the others of which may be saturated, partially unsaturated, or aromatic, for example, dihydronaphthyl, indenyl, indanyl, or tetrahydronaphthyl (tetralinyl). In certain embodiments, aryl may be optionally substituted with one or more substituents Q as described herein.

The term “aralkyl” or “arylalkyl” refers to a monovalent alkyl group substituted with one or more aryl groups. In certain embodiments, the aralkyl has from 7 to 30 (C7-30), from 7 to 20 (C7-20), or from 7 to 16 (C7-16) carbon atoms. Examples of aralkyl groups include, but are not limited to, benzyl, 2-phenylethyl, and 3-phenylpropyl. In certain embodiments, the aralkyl are optionally substituted with one or more substituents Q as described herein.

The term “heteroaryl” refers to a monovalent monocyclic aromatic group or monovalent polycyclic aromatic group that contain at least one aromatic ring, wherein at least one aromatic ring contains one or more heteroatoms independently selected from O, S, N, and P in the ring. A heteroaryl group is bonded to the rest of a molecule through its aromatic ring. Each ring of a heteroaryl group can contain one or two O atoms, one or two S atoms, one to four N atoms, and/or one or two P atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom. In certain embodiments, the heteroaryl has from 5 to 20, from 5 to 15, or from 5 to 10 ring atoms. Examples of monocyclic heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl, and triazolyl. Examples of bicyclic heteroaryl groups include, but are not limited to, benzofuranyl, benzimidazolyl, benzoisoxazolyl, benzopyranyl, benzothiadiazolyl, benzothiazolyl, benzothienyl, benzotriazolyl, benzoxazolyl, furopyridyl, imidazopyridinyl, imidazothiazolyl, indolizinyl, indolyl, indazolyl, isobenzofuranyl, isobenzothienyl, isoindolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxazolopyridinyl, phthalazinyl, pteridinyl, purinyl, pyridopyridyl, pyrrolopyridyl, quinolinyl, quinoxalinyl, quinazolinyl, thiadiazolopyrimidyl, and thienopyridyl. Examples of tricyclic heteroaryl groups include, but are not limited to, acridinyl, benzindolyl, carbazolyl, dibenzofuranyl, perimidinyl, phenanthrolinyl, phenanthridinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and xanthenyl. In certain embodiments, the heteroaryl may also be optionally substituted with one or more substituents Q as described herein as described herein.

The term “heterocyclyl” or “heterocyclic” refers to a monovalent monocyclic non-aromatic ring system or monovalent polycyclic ring system that contains at least one non-aromatic ring, wherein one or more of the non-aromatic ring atoms are heteroatoms independently selected from O, S, N, and P; and the remaining ring atoms are carbon atoms. In certain embodiments, the heterocyclyl or heterocyclic group has from 3 to 20, from 3 to 15, from 3 to 10, from 3 to 8, from 4 to 7, or from 5 to 6 ring atoms. A heterocyclyl group is bonded to the rest of a molecule through its non-aromatic ring. In certain embodiments, the heterocyclyl is a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may be spiro, fused, or bridged, and in which nitrogen or sulfur atoms may be optionally oxidized, nitrogen atoms may be optionally quaternized, and some rings may be partially or fully saturated, or aromatic. The heterocyclyl may be attached to the main structure at any heteroatom or carbon atom which results in the creation of a stable compound. Examples of such heterocyclic groups include, but are not limited to, azepinyl, benzodioxanyl, benzodioxolyl, benzofuranonyl, benzopyranonyl, benzopyranyl, benzotetrahydrofuranyl, benzotetrahydrothienyl, benzothiopyranyl, benzoxazinyl, β-carbolinyl, chromanyl, chromonyl, cinnolinyl, coumarinyl, decahydroisoquinolinyl, dihydrobenzisothiazinyl, dihydrobenzisoxazinyl, dihydrofuryl, dihydroisoindolyl, dihydropyranyl, dihydropyrazolyl, dihydropyrazinyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dioxolanyl, 1,4-dithianyl, furanonyl, imidazolidinyl, imidazolinyl, indolinyl, isobenzotetrahydrofuranyl, isobenzotetrahydrothienyl, isochromanyl, isocoumarinyl, isoindolinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, oxazolidinonyl, oxazolidinyl, oxiranyl, piperazinyl, piperidinyl, 4-piperidonyl, pyrazolidinyl, pyrazolinyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydropyranyl, tetrahydrothienyl, thiamorpholinyl, thiazolidinyl, tetrahydroquinolinyl, and 1,3,5-trithianyl. In certain embodiments, the heterocyclyl may also be optionally substituted with one or more substituents Q as described herein.

The term “halogen”, “halide” or “halo” refers to fluorine, chlorine, bromine, and/or iodine.

The term “optionally substituted” is intended to mean that a group or substituent, such as an alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, cycloalkenyl, aryl, aralkyl, heteroaryl, heteroaryl-C1-6 alkyl, and heterocyclyl group, may be substituted with one or more substituents Q, each of which is independently selected from, e.g., (a) oxo (═O), halo, cyano (—CN), and nitro (—NO2); (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, four, or five, substituents Qa; and (c) —C(O)Ra, —C(O)ORa, —C(O)NRbRc, —C(NRa)NRbRc, —ORa,

—OC(O)Ra, —OC(O)ORa, —OC(O)NRbRc, —OC(═NRa)NRbRc, —OS(O)Ra, —OS(O)2RC, —OS(O)NRbRc, —OS(O)2NRbRc, —NRbRc, —NRaC(O)Rd, —NRaC(O)ORd, —NRaC(O)NRbRc, —NRaC(═NRd)NRbRc, —NRaS(O)Rd, —NRaS(O)2Rd, —NRaS(O)NRbRc, —NRaS(O)2NRbRc, —P(O)RaRd, —P(O)(ORa)Rd, —P(O)(ORa)(ORd), —SRa, —S(O)Ra, —S(O)2R1a, —S(O)NRbRC, and —S(O)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; or (iii) Rb and Rc together with the N atom to which they are attached form heteroaryl or heterocyclyl, optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa. As used herein, all groups that can be substituted are “optionally substituted,” unless otherwise specified.

In one embodiment, each substituent Qa is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; and (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) —C(O)Re, —C(O)ORe, —C(O)NRfRg, —C(NRe)NRfRg, —ORe, —OC(O)Re, —OC(O)ORe, —OC(O)NRfRg, —OC(═NRe)NRfRg, —OS(O)Re, —OS(O)2Re, —OS(O)NRfRg, —OS(O)2NRfRg, —NRfRg, —NReC(O)Rh, —NReC(O)ORh, —NReC(O)NRfRg, —NReC(═NRh)NRfRg, —NReS(O)Rh, —NReS(O)2Rh, —NReS(O)NRfRg, —NReS(O)2NRfRg, —P(O)ReRh, —P(O)(ORe)Rh, —P(O)(ORe)(ORh), —SRe, —S(O)Re, —S(O)2Re, —S(O)NRfRg, and —S(O)2NRfRg; wherein each Re, Rf, Rg, and Rh is independently (i) hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (ii) Rf and Rg together with the N atom to which they are attached form heteroaryl or heterocyclyl.

In certain embodiments, “optically active” and “enantiomerically active” refer to a collection of molecules, which has an enantiomeric excess of no less than about 50%, no less than about 70%, no less than about 80%, no less than about 90%, no less than about 91%, no less than about 92%, no less than about 93%, no less than about 94%, no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%, no less than about 99%, no less than about 99.5%, or no less than about 99.8%. In certain embodiments, the compound comprises about 95% or more of the desired enantiomer and about 5% or less of the less preferred enantiomer based on the total weight of the racemate in question.

In describing an optically active compound, the prefixes R and S are used to denote the absolute configuration of the molecule about its chiral center(s). The (+) and (−) are used to denote the optical rotation of the compound, that is, the direction in which a plane of polarized light is rotated by the optically active compound. The (−) prefix indicates that the compound is levorotatory, that is, the compound rotates the plane of polarized light to the left or counterclockwise. The (+) prefix indicates that the compound is dextrorotatory, that is, the compound rotates the plane of polarized light to the right or clockwise. However, the sign of optical rotation, (+) and (−), is not related to the absolute configuration of the molecule, R and S.

The term “isotopic variant” refers to a compound that contains an unnatural proportion of an isotope at one or more of the atoms that constitute such a compound. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen (H), deuterium (2H), tritium (3H), carbon-11 (11C), carbon-12 (12C), carbon-13 (13C), carbon-14 (14C), nitrogen-13 (13N), nitrogen-14 (14N), nitrogen-15 (15N), oxygen-14 (14O), oxygen-15 (15O), oxygen-16 (16O), oxygen-17 (17O), oxygen-18 (18O), fluorine-17 (17F), fluorine-18 (18F), phosphorus-31 (31P), phosphorus-32 (32P), phosphorus-33 (33P), sulfur-32 (32S), sulfur-33 (33S), sulfur-34 (34S), sulfur-35 (35S), sulfur-36 (36S), chlorine-35 (35C1), chlorine-36 (36Cl), chlorine-37 (37Cl), bromine-79 (79Br), bromine-81 (81Br), iodine-123 (123I), iodine-125 (125I), iodine-127 (127I), iodine-129 (129I), and iodine-131 (131I). In certain embodiments, an “isotopic variant” of a compound is in a stable form, that is, non-radioactive. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen (1H), deuterium (2H), carbon-12 (12C), carbon-13 (13C), nitrogen-14 (14N), nitrogen-15 (15N), oxygen-16 (16O), oxygen-17 (17O), oxygen-18 (18O), fluorine-17 (17F), phosphorus-31 (31P), sulfur-32 (32S), sulfur-33 (33S), sulfur-34 (34S), sulfur-36 (36S), chlorine-35 (35C1), chlorine-37 (37C1), bromine-79 (79Br), bromine-81 (81Br), and iodine-127 (127I). In certain embodiments, an “isotopic variant” of a compound is in an unstable form, that is, radioactive. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, tritium (3H), carbon-11 (11C), carbon-14 (14C), nitrogen-13 (13N), oxygen-14 (14O), oxygen-15 (15O), fluorine-18 (18F), phosphorus-32 (32P), phosphorus-33 (33P), sulfur-35 (35S), chlorine-36 (36Cl), iodine-123 (123I), iodine-125 (125I), iodine-129 (129I), and iodine-131 (131I). It will be understood that, in a compound as provided herein, any hydrogen can be 2H, for example, or any carbon can be 13C, for example, or any nitrogen can be 15N, for example, or any oxygen can be 18O, for example, where feasible according to the judgment of one of skill. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of deuterium (D).

The term “solvate” refers to a complex or aggregate formed by one or more molecules of a solute, e.g., a compound provided herein, and one or more molecules of a solvent, which present in a stoichiometric or non-stoichiometric amount. Suitable solvents include, but are not limited to, water, methanol, ethanol, n-propanol, isopropanol, and acetic acid. In certain embodiments, the solvent is pharmaceutically acceptable. In one embodiment, the complex or aggregate is in a crystalline form. In another embodiment, the complex or aggregate is in a noncrystalline form. Where the solvent is water, the solvate is a hydrate. Examples of hydrates include, but are not limited to, a hemihydrate, monohydrate, dihydrate, trihydrate, tetrahydrate, and pentahydrate.

The phrase “an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof” has the same meaning as the phrase “an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant of the compound referenced therein; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug of the compound referenced therein; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug of an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant of the compound referenced therein.” Compounds

In one embodiment, provided herein is a compound of Formula I:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:

X, Y, and Z are each independently N or CRX, with the proviso that at least two of X, Y, and Z are nitrogen atoms; where RX is hydrogen or C1-6 alkyl;

R1 and R2 are each independently (a) hydrogen, cyano, halo, or nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) —C(O)Ra, —C(O)OR1a, —C(O)NR1bR1c, —C(NR1a)NR1bR1c, —OR1a, —OC(O)R1a, —OC(O)OR1a, —OC(O)NR1bR1c, —OC(═NR1a)NR1bR1c, —OS(O)R1a, —OS(O)2R1a, —OS(O)NR1bR1c, —OS(O)2NR1bR1c, —NR1bR1c, —NR1aC(O)R1d, —NR1aC(O)OR1d, —NR1aC(O)NR1bR1c, —NR1aC(═NR1d)NR1bR1c, —NR1aS(O)R1d, —NR1aS(O)2R1d, —NR1aS(O)NR1bR1c, —NR1aS(O)2NR1bR1c, —SR1a, —S(O)R1a, —S(O)2R1a, —S(O)NR1bR1c, or —S(O)2NR1bR1c; wherein each R1a, R1b, R1c, and R1d is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) R1b and R1c together with the N atom to which they are attached form heterocyclyl;

R3 and R4 are each independently hydrogen or C1-6 alkyl; or R3 and R4 are linked together to form a bond, C1-6 alkylene, C1-6 heteroalkylene, C2-6 alkenylene, or C2-6 heteroalkenylene;

R5a and R5b together with the carbon atom to which they are attached form C3-10 cycloalkyl or heterocyclyl;

R5c is C6-14 aryl, heteroaryl, C7-15 aralkyl, or heteroaryl-C1-C6 alkyl; and

R6 is hydrogen, C1-6 alkyl, —S—C1-6 alkyl, —S(O)—C1-6 alkyl, or —SO2—C1-6 alkyl;

wherein each alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaryl-alkyl, and heterocyclyl in R1, R2, R3, R4, R6, RX, R1a, R1b, R1c, R1d, R5a, R5b, and R5c is optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q, wherein each substituent Q is independently selected from (a) oxo, cyano, halo, and nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; and (c) —C(O)Ra, —C(O)ORa, —C(O)NRbRc, —C(NRa)NRbRc, —ORa, —OC(O)Ra, —OC(O)ORa, —OC(O)NRbRc, —OC(═NRa)NRbRc, —OS(O)Ra, —OS(O)2Ra, —OS(O)NRbRc, —OS(O)2NR1bR1c, —NRbRc, —NRaC(O)Rd, —NRaC(O)ORd, —NRaC(O)NRbRc, —NRaC(═NRd)NRbRc, —NRaS(O)Rd, —NRaS(O)2Rd, —NRaS(O)NRbRc, —NRaS(O)2NRbRc, —SRa, —S(O)Ra, —S(O)2Ra, —S(O)NRbRc, and —S(O)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa;

wherein each Qa is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) —C(O)Re, —C(O)ORe, —C(O)NRfRg, —C(NRe)NRfRg, —ORe, —OC(O)Re, —OC(O)ORe, —OC(O)NRfRg, —OC(═NRe)NRfRg, —OS(O)Re, —OS(O)2Re, —OS(O)NRfRg, —OS(O)2NRfRg, —NRfRg, —NReC(O)Rh, —NReC(O)ORh, —NReC(O)NRfRg, —NReC(═NRh)NRfRg, —NReS(O)Rh, —NReS(O)2Rh, —NReS(O)NRfRg, —NReS(O)2NRfRg, —SRe, —S(O)Re, —S(O)2Re, —S(O)NRfRg, and —S(O)2NRfRg; wherein each Re, Rf, Rg, and Rh is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and Rg together with the N atom to which they are attached form heterocyclyl.

In certain embodiments, the compound provided herein is not 4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-6-morpholino-N-(1-phenylcyclopropyl)-pyrimidin-2-amine. In certain embodiments, in Formula I, when X is CH, and R5a and R5b together with the carbon atom to which they are attached form cyclopropyl, R5c is not phenyl. In certain embodiments, in Formula I, when X is CH, and R5a and R5b together with the carbon atom to which they are attached form C3-10 cycloalkyl, R5c is not phenyl. In certain embodiments, R5a is not phenyl. In certain embodiments, R5a and R5b together with the carbon atom to which they are attached do not form cyclopropyl.

In another embodiment, provided herein is a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:

X, Y, and Z are each independently N or CRX, with the proviso that at least two of X, Y, and Z are nitrogen atoms; where RX is hydrogen or C1-6 alkyl;

R1 and R2 are each independently (a) hydrogen, cyano, halo, or nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1e, —C(NR1a)NR1bR1c, —OR1a, —OC(O)R1a, —OC(O)OR1a, —OC(O)NR1bR1c, —OC(═NR1a)NR1bR1c, —OS(O)R1a, —OS(O)2R1a, —OS(O)NR1bR1c, —OS(O)2NR1bR1c, —NR1bR1c, —NR1aC(O)R1d, —NR1aC(O)OR1d, —NR1aC(O)NR1bR1c, —NR1aC(═NR1d)NR1bR1c, —NR1aS(O)R1d, —NR1aS(O)2R1d, —NR1aS(O)NR1bR1c, —NR1aS(O)2NR1bR1c, —SR1a, —S(O)R1a, —S(O)2R1a, —S(O)NR1bR1c, or —S(O)2NR1bR1c; wherein each R1a, R1b, R1c, and R1d is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) R1b and R1c together with the N atom to which they are attached form heterocyclyl;

R3 and R4 are each independently hydrogen or C1-6 alkyl; or R3 and R4 are linked together to form a bond, C1-6 alkylene, C1-6 heteroalkylene, C2-6 alkenylene, or C2-6 heteroalkenylene;

R5a and R5b together with the carbon atom to which they are attached form C3-10 cycloalkyl or heterocyclyl;

R5c is C6-14 aryl or heteroaryl; and

R6 is hydrogen, C1-6 alkyl, —S—C1-6 alkyl, —S(O)—C1-6 alkyl, or —SO2—C1-6 alkyl;

wherein each alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl in R1, R2, R3, R4, R6, RX, R1a, R1b, R1c, R1d, R5a, R5b, and R5c is optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q, wherein each substituent Q is independently selected from (a) oxo, cyano, halo, and nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; and (c) —C(O)Ra, —C(O)ORa, —C(O)NR1bR1c, —C(NRa)NRbRc, —ORa, —OC(O)Ra, —OC(O)ORa, —OC(O)NRbRc, —OC(═NRa)NRbRc, —OS(O)Ra, —OS(O)2R1a, —OS(O)NRbRc, —OS(O)2NRbRc, —NRbRc, —NRaC(O)Rd, —NRaC(O)ORd, —NRaC(O)NRbRc, —NRaC(═NRd)NR1bR1c, —NRaS(O)Rd, —NRaS(O)2Rd, —NRaS(O)NRbRc, —NRaS(O)2NRbRc, —SRa, —S(O)Ra, —S(O)2R1a, —S(O)NRbRc, and —S(O)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa;

wherein each Qa is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) —C(O)Rc, —C(O)ORc, —C(O)NRfRg, —C(NRe)NRfRg, —ORe, —OC(O)Re, —OC(O)ORe, —OC(O)NRfRg, —OC(═NRe)NRfRh, —OS(O)Re, —OS(O)2Re, —OS(O)NRfRg, —OS(O)2NRfRg, —NRfRg, —NReC(O)Rh, —NReC(O)ORh, —NReC(O)NRfRg, —NReC(═NRh)NRfRg, —NReS(O)Rh, —NReS(O)2Rh, —NReS(O)NRfRg, —NReS(O)2NRfRg, —SRe, —S(O)Re, —S(O)2Re, —S(O)NRfRg, and —S(O)2NRfRg; wherein each Re, Rf, Rg, and Rh is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and Rg together with the N atom to which they are attached form heterocyclyl.

In one embodiment, the compound of Formula I has the structure of Formula Ia:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R5c, X, Y, and Z are each as defined herein.

In another embodiment, the compound of Formula I has the structure of Formula Ib:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R5c, X, Y, and Z are each as defined herein.

In yet another embodiment, provided herein is a compound of Formula II:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, and R5c are each as defined herein.

In one embodiment, the compound of Formula II has the structure of Formula IIa:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, and R5c are each as defined herein.

In another embodiment, the compound of Formula II has the structure of Formula IIb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, and R5c are each as defined herein.

In yet another embodiment, provided herein is a compound of Formula III:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:

R7a, R7b, R7c, R7d, and R7e are each independently (a) hydrogen, cyano, halo, or nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more substituents Q; or (c) —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, —C(NR1a)NR1bR1c, —OR1a, —OC(O)R1a, —OC(O)OR1a, —OC(O)NR1bR1c, —OC(═NR1a)NR1bR1c, —OS(O)R1a, —OS(O)2R1a, —OS(O)NR1bR1c, —OS(O)2NR1bR1c, —NR1bR1c, —NR1aC(O)R1d, —NR1aC(O)OR1d, —NR1aC(O)NR1bR1c, —NR1aC(═NR1d)NR1bR1c, —NR1aS(O)R1d, —NR1aS(O)2R1d, —NR1aS(O)NR1bR1c, —NR1aS(O)2NR1bR1c, —SR1a, —S(O)R1a, —S(O)2R1a, —S(O)NR1bR1c, or —S(O)2NR1bR1c;

two of R7a, R7b, R7c, R7d, and R7e that are adjacent to each other form C3-10 cycloalkenyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q; and

R1, R2, R3, R4, R6, R1a, R1b, R1c, R1d, R5a, R5b, X, Y, and Z are each as defined herein.

In one embodiment, the compound of Formula III has the structure of Formula IIIa:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, R7e, X, Y, and Z are each as defined herein.

In another embodiment, the compound of Formula III has the structure of Formula IIIb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, R7e, X, Y, and Z are each as defined herein.

In yet another embodiment, provided herein is a compound of Formula IV:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, and R7e are each as defined herein.

In one embodiment, the compound of Formula IV has the structure of Formula IVa:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, and R7e are each as defined herein.

In another embodiment, the compound of Formula IV has the structure of Formula IVb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, and R7e are each as defined herein.

In yet another embodiment, provided herein is a compound of Formula V:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:

V is a bond, —(CH2)r—, —O(CH2)r—, —S(CH2)r, or —N(R8)(CH2)r—;

each R8 is independently (a) hydrogen; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more substituents Q; or (c) —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, —C(NR1a)NR1bR1c, —OR1a, —OC(O)R1a, —OC(O)OR1a, —OC(O)NR1bR1c, —OC(═NR1a)NR1bR1c, —OS(O)R1a, —OS(O)2R1a, —OS(O)NR1bR1c, —OS(O)2NR1bR1c, —NR1bR1c, —NR1aC(O)R1d, —NR1aC(O)OR1d, —NR1aC(O)NR1bR1c, —NR1aC(═NR1d)NR1bR1c, —NR1aS(O)R1d, —NR1aS(O)2R1d, —NR1aS(O)NR1bR1c, —NR1aS(O)2NR1bR1c, —S(O)Ra, —S(O)2R1a, —S(O)NR1bR1c, or —S(O)2NR1bR1c;

m and r are each an integer of 0, 1, or 2;

n is an integer of 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and

R1, R2, R3, R4, R6, R1a, R1b, R1c, R1d, R5c, X, Y, and Z are each as defined herein.

In one embodiment, the compound of Formula V has the structure of Formula Va:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R5c, V, X, Y, Z, m, and n are each as defined herein.

In another embodiment, the compound of Formula V has the structure of Formula Vb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R5c, V, X, Y, Z, m, and n are each as defined herein.

In yet another embodiment, provided herein is a compound of Formula VI:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R5c, V, m, and n are each as defined herein.

In one embodiment, the compound of Formula VI has the structure of Formula VIa:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R5c, V, m, and n are each as defined herein.

In another embodiment, the compound of Formula VI has the structure of Formula VIb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R5c, V, m, and n are each as defined herein.

In yet another embodiment, provided herein is a compound of Formula VII:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, X, Y, Z, m, and n are each as defined herein.

In one embodiment, the compound of Formula VII has the structure of Formula VIIa:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, X, Y, Z, m, and n are each as defined herein.

In another embodiment, the compound of Formula VII has the structure of Formula VIIb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, X, Y, Z, m, and n are each as defined herein.

In yet another embodiment, provided herein is a compound of Formula VIII:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, m, and n are each as defined herein.

In one embodiment, the compound of Formula VIII has the structure of Formula VIIIa:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, m, and n are each as defined herein.

In another embodiment, the compound of Formula VIII has the structure of Formula VIIIb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, m, and n are each as defined herein.

In yet another embodiment, provided herein is a compound of Formula IX:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, R7e, X, Y, and Z are each as defined herein; and k is an integer of 1, 2, 3, 4, 5, or 6. In certain embodiments, k is an integer of 1.

In one embodiment, the compound of Formula IX has the structure of Formula IXa:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, R7e, X, Y, Z, and k are each as defined herein. In certain embodiments, k is an integer of 1.

In another embodiment, the compound of Formula IX has the structure of Formula IXb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, R7e, X, Y, Z, and k are each as defined herein. In certain embodiments, k is an integer of 1.

In yet another embodiment, provided herein is a compound of Formula X:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, R7e, and k are each as defined herein. In certain embodiments, k is an integer of 1.

In one embodiment, the compound of Formula X has the structure of Formula Xa:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, R7e, and k are each as defined herein. In certain embodiments, k is an integer of 1.

In another embodiment, the compound of Formula X has the structure of Formula Xb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R5a, R5b, R7a, R7b, R7c, R7d, R7e, and k are each as defined herein. In certain embodiments, k is an integer of 1.

In yet another embodiment, provided herein is a compound of Formula XI:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, X, Y, Z, k, m, and n are each as defined herein. In certain embodiments, k is an integer of 1.

In one embodiment, the compound of Formula XI has the structure of Formula

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, X, Y, Z, k, m, and n are each as defined herein. In certain embodiments, k is an integer of 1.

In another embodiment, the compound of Formula XI has the structure of Formula XIb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, X, Y, Z, k, m, and n are each as defined herein. In certain embodiments, k is an integer of 1.

In still another embodiment, provided herein is a compound of Formula XII:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, k, m, and n are each as defined herein. In certain embodiments, k is an integer of 1.

In one embodiment, the compound of Formula XII has the structure of Formula XIIa:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8R7a, R7b, R7c, R7d, R7e, V, k, m, and n are each as defined herein. In certain embodiments, k is an integer of 1.

In another embodiment, the compound of Formula XII has the structure of Formula XIIb:

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein R1, R2, R3, R4, R6, R8, R7a, R7b, R7c, R7d, R7e, V, k, m, and n are each as defined herein. In certain embodiments, k is an integer of 1.

The groups, R1, R2, R3, R4, R6, R8, R5a, R5b, R5c, R7a, R7b, R7c, R7e, V, X, Y, Z, k, m, n, and r in Formulae provided herein, e.g., Formulae I to XII, la to XIIa, and Ib to XIIb, are further defined in the embodiments described herein. All combinations of the embodiments provided herein for such groups are within the scope of this disclosure.

In certain embodiments, R1 is hydrogen. In certain embodiments, R1 is cyano. In certain embodiments, R1 is halo. In certain embodiments, R1 is fluoro, chloro, bromo, or iodo. In certain embodiments, R1 is nitro. In certain embodiments, R1 is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is C2-6 alkenyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is C2-6 alkynyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is C3-10 cycloalkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is C6-14 aryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is C7-15 aralkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is heteroaryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is heterocyclyl, optionally substituted with one or more substituents Q as described herein.

In certain embodiments, R1 is —C(O)R1a, wherein R1a is as defined herein. In certain embodiments, R1 is —C(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R1 is —C(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R1 is —C(NR1a)NR1bR1a, wherein R1a, R1b, and R1C are each as defined herein. In certain embodiments, R1 is —OR1a, wherein R1a is as defined herein. In certain embodiments, R1 is —O—C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is methoxy, ethoxy, propoxy, isopropoxy, or 3-dimethylaminopropoxy. In certain embodiments, R1 is —OC(O)R1a, wherein R1a is as defined herein. In certain embodiments, R1 is —OC(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R1 is —OC(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R1 is —OC(═NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R1 is —OS(O)R1a, wherein R1a is as defined herein. In certain embodiments, R1 is —OS(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R1 is —OS(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R1 is —OS(O)2NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R1 is —NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R1 is —NR1aC(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R1 is —NR1aC(O)OR1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R1 is —NR1aC(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R1 is —NR1aC(═NRd)NR1bR1c, wherein R1a, R1b, R1c, and R1d are each as defined herein. In certain embodiments, R1 is —NR1aS(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R1 is —NR1aS(O)2R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R1 is —NR1aS(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R1 is —NR1aS(O)2NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R1 is —SR1a, wherein R1a is as defined herein. In certain embodiments, R1 is —S(O)R1a, wherein R1a is as defined herein. In certain embodiments, R1 is —S(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R1 is —S(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R1 is —S(O)2NR1bR1a; wherein R1b and R1c are each as defined herein.

In certain embodiments, R2 is hydrogen. In certain embodiments, R2 is cyano. In certain embodiments, R2 is halo. In certain embodiments, R2 is fluoro, chloro, bromo, or iodo. In certain embodiments, R2 is nitro. In certain embodiments, R2 is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R2 is C2-6 alkenyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R2 is C2-6 alkynyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R2 is C3-7 cycloalkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R2 is C6-14 aryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R2 is C7-15 aralkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R2 is heteroaryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R2 is heterocyclyl, optionally substituted with one or more substituents Q as described herein.

In certain embodiments, R2 is —C(O)R1a, wherein R1a is as defined herein. In certain embodiments, R2 is —C(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R2 is —C(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R2 is —C(NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R2 is —OR1a, wherein R1a is as defined herein. In certain embodiments, R1 is —O—C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is methoxy, ethoxy, propoxy, isopropoxy, or 3-dimethylaminopropoxy. In certain embodiments, R2 is —OC(O)R1a, wherein R1a is as defined herein. In certain embodiments, R2 is —OC(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R2 is —OC(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R2 is —OC(═NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R2 is —OS(O)R1a, wherein R1a is as defined herein. In certain embodiments, R2 is —OS(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R2 is —OS(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R2 is —OS(O)2NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R2 is —NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R2 is amino (—NH2). In certain embodiments, R2 is —NR1aC(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R2 is —NR1aC(O)OR1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R2 is —NR1aC(O)NR1bR1c, wherein R1a, R1b, and Ric are each as defined herein. In certain embodiments, R2 is —NR1aC(═NR1d)NR1bR1c, wherein R1a, R1b, R1c, and R1d are each as defined herein. In certain embodiments, R2 is —NR1aS(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R2 is —NR1aS(O)2R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R2 is —NR1aS(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R2 is —NR1aS(O)2NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R2 is —SR1a, wherein R1a is as defined herein. In certain embodiments, R2 is —S(O)R1a, wherein R1a is as defined herein. In certain embodiments, R2 is —S(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R2 is —S(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R2 is —S(O)2NR1bR1c; wherein R1b and R1c are each as defined herein.

In certain embodiments, R3 is hydrogen. In certain embodiments, R3 is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R3 is hydrogen, methyl, ethyl, or propyl (e.g., n-propyl, isopropyl, or 2-isopropyl).

In certain embodiments, R4 is hydrogen. In certain embodiments, R4 is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R4 is hydrogen, methyl, ethyl, or propyl (e.g., n-propyl, isopropyl, or 2-isopropyl).

In certain embodiments, R3 and R4 are linked together to form a bond. In certain embodiments, R3 and R4 are linked together to form C1-6 alkylene, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R3 and R4 are linked together to form methylene, ethylene, or propylene, each optionally substituted with one or more substituents Q as described herein. In certain embodiments, R3 and R4 are linked together to form C1-6 heteroalkylene, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R3 and R4 are linked together to form C2-6 alkenylene, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R3 and R4 are linked together to form C2-6 heteroalkenylene, optionally substituted with one or more substituents Q as described herein.

In certain embodiments, R6 is hydrogen. In certain embodiments, R6 is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R6 is C1-6 alkyl, optionally substituted with one or more, in one embodiment, one, two, or three, halo. In certain embodiments, R6 is C1-6 alkyl, optionally substituted with one or more, in one embodiment, one, two, or three, fluoro. In certain embodiments, R6 is methyl, fluoromethyl, difluoromethyl, or trifluoromethyl. In certain embodiments, R6 is difluoromethyl. In certain embodiments, R6 is —S—C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q as described herein. In certain embodiments, R6 is —S(O)—C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q as described herein. In certain embodiments, R6 is —SO2—C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q as described herein.

In certain embodiments, R8 is hydrogen. In certain embodiments, R8 is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R8 is C2-6 alkenyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R8 is C2-6 alkynyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R8 is C3-7 cycloalkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R8 is C6-14 aryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R8 is C7-15 aralkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R8 is heteroaryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R8 is heterocyclyl, optionally substituted with one or more substituents Q as described herein.

In certain embodiments, R8 is —C(O)R1a, wherein R1a is as defined herein. In certain embodiments, R8 is —C(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R8 is —C(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R8 is —C(NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R8 is —OR1a, wherein R1a is as defined herein. In certain embodiments, R8 is —OC(O)R1a, wherein R11 is as defined herein. In certain embodiments, R8 is —OC(O)OR1a, wherein R11 is as defined herein. In certain embodiments, R8 is —OC(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R8 is —OC(═NR1a)NR1bR1a, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R8 is —OS(O)R1a, wherein R1a is as defined herein. In certain embodiments, R8 is —OS(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R8 is —OS(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R8 is —OS(O)2NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R8 is —NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R8 is amino (—NH2). In certain embodiments, R8 is —NR1aC(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R8 is —NR1aC(O)OR1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R8 is —NR1aC(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R8 is —NRaC(═NR1d)NR1bR1c wherein R1a, R1b, R1c, and R1d are each as defined herein. In certain embodiments, R8 is —NR1aS(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R8 is —NR1aS(O)2R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R8 is —NR1aS(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R8 is —NR1S(O)2NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R8 is —S(O)R1a, wherein R1a is as defined herein. In certain embodiments, R1 is —S(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R8 is —S(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R8 is —S(O)2NR1bR1c; wherein R1b and R1c are each as defined herein.

In certain embodiments, R5a and R5b together with the C atom to which they are attached form C3-10 cycloalkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5a and R5b together with the C atom to which they are attached form cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl, each optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5a and R5b together with the C atom to which they are attached form heterocyclyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5a and R5b together with the C atom to which they are attached form monocyclic heterocyclyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5a and R5b together with the C atom to which they are attached form 5- or 6-membered heterocyclyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5a and R5b together with the C atom to which they are attached form piperidinyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5a and R5b together with the C atom to which they are attached form piperidin-4-yl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5a and R5b together with the C atom to which they are attached form N-methyl-piperidin-4-yl.

In certain embodiments, R5c is C6-14 aryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5c is phenyl or naphthyl, each optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5c is phenyl, naphtha-1-yl, or naphtha-2-yl, each optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5c is phenyl, 4-chlorophenyl, 4-methoxyphenyl, or naphtha-2-yl. In certain embodiments, R5c is heteroaryl, optionally substituted with one or more substituents as described herein. In certain embodiments, R5c is moncyclic heteroaryl, optionally substituted with one or more substituents as described herein. In certain embodiments, R5c is 5- or 6-membered heteroaryl, optionally substituted with one or more substituents as described herein. In certain embodiments, R5c is bicyclic heteroaryl, optionally substituted with one or more substituents as described herein.

In certain embodiments, R5c is C7-15 aralkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5c is benzyl, 2-phenylethyl, 3-phenylpropal, or 4-phenylbutyl, each optionally substituted with one or more substituents Q as described herein. In certain embodiments, R5c is benzyl, fluorobenzyl, chlorobenzyl, bromobenzyl, or methylbenzyl. In certain embodiments, R5c is benzyl, 2-fluorobenzyl, 3-fluorobenzyl, 4-fluorobenzyl, 2-chlorobenzyl, 3-chlorobenzyl, 4-chlorobenzyl, 2-bromobenzyl, 3-bromobenzyl, 4-bromobenzyl, 2-methylbenzyl, 3-methylbenzyl, or 4-methylbenzyl. In certain embodiments, R5c is heteroaryl-C1-6 alkyl, optionally substituted with one or more substituents as described herein.

In certain embodiments, R7a is hydrogen. In certain embodiments, R7a is cyano. In certain embodiments, R7a is halo. In certain embodiments, R7a is fluoro, chloro, bromo, or iodo. In certain embodiments, R7a is nitro. In certain embodiments, R7a is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7a is C2-6 alkenyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7a is C2-6 alkynyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7a is C3-7 cycloalkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7a is C6-14 aryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7a is C7-15 aralkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7a is heteroaryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7a is heterocyclyl, optionally substituted with one or more substituents Q as described herein.

In certain embodiments, R7a is —C(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7a is —C(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R7a is —C(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7a is —C(NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7a is —ORa, wherein R1a is as defined herein. In certain embodiments, R1 is —O—C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is methoxy, ethoxy, propoxy, isopropoxy, or 3-dimethylaminopropoxy. In certain embodiments, R7a is —OC(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7a is —OC(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R7a is —OC(O)NRO1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7a is —OC(═NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7a is —OS(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7a is —OS(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R7a is —OS(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7a is —OS(O)2NR1bR1c wherein R1b and R1c are each as defined herein. In certain embodiments, R7a is —NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7a is amino (—NH2). In certain embodiments, R7a is —NR1aC(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7a is —NR1aC(O)OR1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7a is —NR1aC(O)NR(1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7a is —NR1aC(═NRdNR1bR1c, wherein R1a, R1b, R1c, and R1d are each as defined herein. In certain embodiments, R7a is —NR1aS(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7a is —NR1aS(O)2R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7a is —NR1aS(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7a is —NR1aS(O)2NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7a is —SR1a, wherein R1a is as defined herein. In certain embodiments, R7a is —S(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7a is —S(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R7a is —S(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7a is —S(O)2NR1bR1c; wherein R1b and R1c are each as defined herein.

In certain embodiments, R7b is hydrogen. In certain embodiments, R7b is cyano. In certain embodiments, R7b is halo. In certain embodiments, R7b is fluoro, chloro, bromo, or iodo. In certain embodiments, R7b is nitro. In certain embodiments, R7b is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7b is C2-6 alkenyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7b is C2-6 alkynyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7b is C3-7 cycloalkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7b is C6-14 aryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7b is C7-15 aralkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7b is heteroaryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7b is heterocyclyl, optionally substituted with one or more substituents Q as described herein.

In certain embodiments, R7b is —C(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7b is —C(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R7b is —C(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7b is —C(NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7b is —OR1a, wherein R1a is as defined herein. In certain embodiments, R1 is —O—C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is methoxy, ethoxy, propoxy, isopropoxy, or 3-dimethylaminopropoxy. In certain embodiments, R7b is —OC(O)R1a, wherein Ra is as defined herein. In certain embodiments, R7b is —OC(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R7b is —OC(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7b is —OC(═NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7b is —OS(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7b is —OS(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R7b is —OS(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7b is —OS(O)2NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7b is —NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7b is amino (—NH2). In certain embodiments, R7b is —NR1aC(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7b is —NR1aC(O)OR1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7b is —NR1aC(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7b is —NR1aC(═NR1d)NR1bR1c, wherein R1a, R1b, Rc, and R1d are each as defined herein. In certain embodiments, R7b is —NR1aS(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7b is —NR1aS(O)2R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7b is —NRaS(O)2R1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7b is —NR1aS(O)2NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7b is —SR1a, wherein R1a is as defined herein. In certain embodiments, R7b is —S(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7b is —S(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R7b is —S(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7b is —S(O)2NR1bR1a; wherein R1b and R1c are each as defined herein.

In certain embodiments, R7c is hydrogen. In certain embodiments, R7c is cyano. In certain embodiments, R7c is halo. In certain embodiments, R7c is fluoro, chloro, bromo, or iodo. In certain embodiments, R7c is nitro. In certain embodiments, R7c is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7c is C2-6 alkenyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7c is C2-6 alkynyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7c is C3-7 cycloalkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7c is C6-14 aryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7c is C7-15 aralkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7c is heteroaryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7c is heterocyclyl, optionally substituted with one or more substituents Q as described herein.

In certain embodiments, R7c is —C(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7c is —C(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R7c is —C(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7c is —C(NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7c is —OR1a, wherein R1a is as defined herein. In certain embodiments, R1 is —O—C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is methoxy, ethoxy, propoxy, isopropoxy, or 3-dimethylaminopropoxy. In certain embodiments, R7c is —OC(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7c is —OC(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R7c is —OC(O)NRO1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7c is —OC(═NR1a)NR1bR1c, wherein R1a, Rb, and R1c are each as defined herein. In certain embodiments, R7 is —OS(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7c is —OS(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R7c is —OS(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7c is —OS(O)2NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7c is —NRbRc, wherein R1b and R1c are each as defined herein. In certain embodiments, R7c is amino (—NH2). In certain embodiments, R7c is —NR1aC(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7c is —NR1aC(O)OR1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7c is —NR1aC(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7c is —NR1aC(═NR1d)NR1bR1c, wherein R1a, R1b, Rc, and R1d are each as defined herein. In certain embodiments, R7c is —NR1aS(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7c is —NR1aS(O)2R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7c is —NR1aS(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7c is —NR1aS(O)2NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7c is —SR1a, wherein R1a is as defined herein. In certain embodiments, R7c is —S(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7 is —S(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R7 is —S(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7c is —S(O)2NR1bR1c; wherein R1b and R1c are each as defined herein.

In certain embodiments, R7d is hydrogen. In certain embodiments, R7d is cyano. In certain embodiments, R7d is halo. In certain embodiments, R7d is fluoro, chloro, bromo, or iodo. In certain embodiments, R7d is nitro. In certain embodiments, R7d is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7d is C2-6 alkenyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7d is C2-6 alkynyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7d is C3-7 cycloalkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7d is C6-14 aryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7d is C7-15 aralkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7d is heteroaryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7d is heterocyclyl, optionally substituted with one or more substituents Q as described herein.

In certain embodiments, R7d is —C(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7d is —C(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R7d is —C(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7d is —C(NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7d is —OR1a, wherein R1a is as defined herein. In certain embodiments, R1 is —O—C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is methoxy, ethoxy, propoxy, isopropoxy, or 3-dimethylaminopropoxy. In certain embodiments, R7d is —OC(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7d is —OC(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R7d is —OC(O)NRO1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7d is —OC(═NR1a)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7d is —OS(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7d is —OS(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R7d is —OS(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7d is —OS(O)2NR1bR1c wherein R1b and R1c are each as defined herein. In certain embodiments, R7d is —NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7d is amino (—NH2). In certain embodiments, R7d is —NR1aC(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7d is —NR1aC(O)OR1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7d is —NR1aC(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7d is —NR1aC(═NR1d)NR1bR1c, wherein R1a, R1b, R1c, and R1d are each as defined herein. In certain embodiments, R7d is —NR1aS(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7d is —NRaS(O)2R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7d is —NR1aS(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7d is —NR1aS(O)2NRObR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7d is —SR1a, wherein R1a is as defined herein. In certain embodiments, R7d is —S(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7d is —S(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R7d is —S(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7d is —S(O)2NR1bR1c; wherein R1b and R1c are each as defined herein.

In certain embodiments, R7e is hydrogen. In certain embodiments, R7e is cyano. In certain embodiments, R7e is halo. In certain embodiments, R7e is fluoro, chloro, bromo, or iodo. In certain embodiments, R7e is nitro. In certain embodiments, R7e is C1-6 alkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7e is C2-6 alkenyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7e is C2-6 alkynyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7e is C3-7 cycloalkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7e is C6-14 aryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7e is C7-15 aralkyl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7e is heteroaryl, optionally substituted with one or more substituents Q as described herein. In certain embodiments, R7e is heterocyclyl, optionally substituted with one or more substituents Q as described herein.

In certain embodiments, R7e is —C(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7e is —C(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R7e is —C(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7e is —C(NR1a)NR1bR1cC, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7e is —OR1a, wherein R1a is as defined herein. In certain embodiments, R1 is —O—C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q as described herein. In certain embodiments, R1 is methoxy, ethoxy, propoxy, isopropoxy, or 3-dimethylaminopropoxy. In certain embodiments, R7e is —OC(O)R1a, wherein R1 is as defined herein. In certain embodiments, R7e is —OC(O)OR1a, wherein R1a is as defined herein. In certain embodiments, R7e is —OC(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7e is —OC(═NR1a)NR1bR1c, wherein R1a, R1b, and R1 are each as defined herein. In certain embodiments, R7e is —OS(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7e is —OS(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R7e is —OS(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7e is —OS(O)2NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7e is —NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7e is amino (—NH2). In certain embodiments, R7e is —NR1aC(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7e is —NR1aC(O)OR1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7e is —NR1aC(O)NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7e is —NR1aC(═NR1d)NR1bR1c, wherein R1a, R1b, R1c, and R1d are each as defined herein. In certain embodiments, R7e is —NR1aS(O)R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7e is —NR1aS(O)2R1d, wherein R1a and R1d are each as defined herein. In certain embodiments, R7e is —NR1aS(O)2R1bRc, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7e is —NR1aS(O)2NR1bR1c, wherein R1a, R1b, and R1c are each as defined herein. In certain embodiments, R7e is —SR1a, wherein R1a is as defined herein. In certain embodiments, R7e is —S(O)R1a, wherein R1a is as defined herein. In certain embodiments, R7e is —S(O)2R1a, wherein R1a is as defined herein. In certain embodiments, R7e is —S(O)NR1bR1c, wherein R1b and R1c are each as defined herein. In certain embodiments, R7e is —S(O)2NR1bR1c; wherein R1b and R1c are each as defined herein.

In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form C3-10 cycloalkenyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form C3-10 cycloalkenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form cyclohexenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form C6-14 aryl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form phenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form monocyclic heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form 5- or 6-membered heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form bicyclic heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form monocyclic heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form 5- or 6-membered heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7a and R7b together with the carbon atoms to which they are attached form bicyclic heterocyclyl, optionally substituted with one or more substituents Q.

In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form C3-10 cycloalkenyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form C3-10 cycloalkenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form cyclohexenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form C6-14 aryl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form phenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form monocyclic heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form 5- or 6-membered heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form bicyclic heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form monocyclic heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form 5- or 6-membered heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7b and R7c together with the carbon atoms to which they are attached form bicyclic heterocyclyl, optionally substituted with one or more substituents Q.

In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form C3-10 cycloalkenyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form C3-10 cycloalkenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form cyclohexenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7e and R7d together with the carbon atoms to which they are attached form C6-14 aryl, optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form phenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form monocyclic heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form 5- or 6-membered heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form bicyclic heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form monocyclic heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form 5- or 6-membered heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7c and R7d together with the carbon atoms to which they are attached form bicyclic heterocyclyl, optionally substituted with one or more substituents Q.

In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form C3-10 cycloalkenyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form C3-10 cycloalkenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form cyclohexenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form C6-14 aryl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form phenyl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form monocyclic heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form 5- or 6-membered heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form bicyclic heteroaryl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form monocyclic heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form 5- or 6-membered heterocyclyl, optionally substituted with one or more substituents Q. In certain embodiments, R7d and R7e together with the carbon atoms to which they are attached form bicyclic heterocyclyl, optionally substituted with one or more substituents Q

In certain embodiments, V is a bond. In certain embodiments, V is —(CH2)r—, wherein r is as defined herein and the alkylene, i.e., —(CH2)r—, is optionally substituted with one or more substituents R8 as defined herein. In certain embodiments, V is —O(CH2)r—, wherein r is as defined herein and the alkylene is optionally substituted with one or more substituents R8 as defined herein. In certain embodiments, V is —O—. In certain embodiments, V is —S(CH2)r—, wherein r is as defined herein and the alkylene is optionally substituted with one or more substituents R8 as defined herein. In certain embodiments, V is —S—. In certain embodiments, V is —N(Rs)(CH2)r—, wherein R8 and r are each as defined herein and the alkylene is optionally substituted with one or more substituents R8 as defined herein. In certain embodiments, V is —N(Rs)—, wherein R8 is as defined herein. In certain embodiments, V is —N(C1-6 alkyl)-, wherein the alkyl is optionally substituted with one or more substituents R8 as defined herein. In certain embodiments, V is —N(CH3)—.

In certain embodiments, X is N In certain embodiments, X is CRX, wherein RX is as defined herein. In certain embodiments, X is CH.

In certain embodiments, Y is N In certain embodiments, Y is CRX, wherein RX is as defined herein. In certain embodiments, Y is CH.

In certain embodiments, Z is N In certain embodiments, Z is CRX, wherein RX is as defined herein. In certain embodiments, Z is CH.

In certain embodiments, X, Y, and Z are N. In certain embodiments, X and Y are N, and Z is CH. In certain embodiments, X and Z are N, and Y is CH. In certain embodiments, Y and Z are N, and X is CH.

In certain embodiments, k is an integer of 1. In certain embodiments, k is an integer of 2. In certain embodiments, k is an integer of 3. In certain embodiments, k is an integer of 4. In certain embodiments, k is an integer of 5. In certain embodiments, k is an integer of 6.

In certain embodiments, m is an integer of 0. In certain embodiments, m is an integer of 1. In certain embodiments, m is an integer of 2.

In certain embodiments, n is an integer of 0. In certain embodiments, n is an integer of 1. In certain embodiments, n is an integer of 2. In certain embodiments, n is an integer of 3. In certain embodiments, n is an integer of 4. In certain embodiments, n is an integer of 5. In certain embodiments, n is an integer of 6. In certain embodiments, n is an integer of 7. In certain embodiments, n is an integer of 8. In certain embodiments, n is an integer of 9. In certain embodiments, n is an integer of 10.

In certain embodiments, r is an integer of 0. In certain embodiments, r is an integer of 1. In certain embodiments, r is an integer of 2.

In one embodiment, provided herein is a compound selected from:

and enantiomers, mixtures of enantiomers, mixtures of two or more diastereomers, and isotopic variants thereof; and pharmaceutically acceptable salts, solvates, hydrates, and prodrugs thereof.

The compounds provided herein are intended to encompass all possible stereoisomers, unless a particular stereochemistry is specified. Where the compound provided herein contains an alkenyl or alkenylene group, the compound may exist as one or mixture of geometric cis/trans (or Z/E) isomers. Where structural isomers are interconvertible, the compound may exist as a single tautomer or a mixture of tautomers. This can take the form of proton tautomerism in the compound that contains, for example, an imino, keto, or oxime group; or so-called valence tautomerism in the compound that contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.

The compounds provided herein may be enantiomerically pure, such as a single enantiomer or a single diastereomer, or be stereoisomeric mixtures, such as a mixture of enantiomers, e.g., a racemic mixture of two enantiomers; or a mixture of two or more diastereomers. As such, one of skill in the art will recognize that administration of a compound in its (R) form is equivalent, for compounds that undergo epimerization in vivo, to administration of the compound in its (S) form. Conventional techniques for the preparation/isolation of individual enantiomers include synthesis from a suitable optically pure precursor, asymmetric synthesis from achiral starting materials, or resolution of an enantiomeric mixture, for example, chiral chromatography, recrystallization, resolution, diastereomeric salt formation, or derivatization into diastereomeric adducts followed by separation.

When the compound provided herein contains an acidic or basic moiety, it may also be provided as a pharmaceutically acceptable salt (See, Berge et al., J. Pharm. Sci. 1977, 66, 1-19; and “Handbook of Pharmaceutical Salts, Properties, and Use,” Stahl and Wermuth, Ed.; Wiley-VCH and VHCA, Zurich, 2002).

Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, (+)-camphoric acid, camphorsulfonic acid, (+)-(1S)-camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, D-gluconic acid, D-glucuronic acid, L-glutamic acid, α-oxoglutaric acid, glycolic acid, hippuric acid, hydrobromic acid, hydrochloric acid, hydroiodic acid, (+)-L-lactic acid, (+)-DL-lactic acid, lactobionic acid, lauric acid, maleic acid, (−)-L-malic acid, malonic acid, (+)-DL-mandelic acid, methanesulfonic acid, naphthalene-2-sulfonic acid, naphthalene-1,5-disulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, nitric acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, perchloric acid, phosphoric acid, L-pyroglutamic acid, saccharic acid, salicylic acid, 4-amino-salicylic acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid, undecylenic acid, and valeric acid.

Suitable bases for use in the preparation of pharmaceutically acceptable salts, including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium hydroxide, potassium hydroxide, zinc hydroxide, or sodium hydroxide; and organic bases, such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol, diethanolamine, diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2-(diethylamino)-ethanol, ethanolamine, ethylamine, ethylenediamine, isopropylamine, N-methyl-glucamine, hydrabamine, 1H-imidazole, L-lysine, morpholine, 4-(2-hydroxyethyl)-morpholine, methylamine, piperidine, piperazine, propylamine, pyrrolidine, 1-(2-hydroxyethyl)-pyrrolidine, pyridine, quinuclidine, quinoline, isoquinoline, secondary amines, triethanolamine, trimethylamine, triethylamine, N-methyl-D-glucamine, 2-amino-2-(hydroxymethyl)-1,3-propanediol, and tromethamine.

The compound provided herein may also be provided as a prodrug, which is a functional derivative of the compound, for example, of Formula I, and is readily convertible into the parent compound in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have enhanced solubility in pharmaceutical compositions over the parent compound. A prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis. See Harper, Progress in Drug Research 1962, 4, 221-294; Morozowich et al. in “Design of Biopharmaceutical Properties through Prodrugs and Analogs,” Roche Ed., APHA Acad. Pharm. Sci. 1977; “Bioreversible Carriers in Drug in Drug Design, Theory and Application,” Roche Ed., APHA Acad. Pharm. Sci. 1987; “Design of Prodrugs,” Bundgaard, Elsevier, 1985; Wang et al., Curr. Pharm. Design 1999, 5, 265-287; Pauletti et al., Adv. Drug. Delivery Rev. 1997, 27, 235-256; Mizen et al., Pharm. Biotech. 1998, 11, 345-365; Gaignault et al., Pract. Med. Chem. 1996, 671-696; Asgharnejad in “Transport Processes in Pharmaceutical Systems,” Amidon et al., Ed., Marcell Dekker, 185-218, 2000; Balant et al., Eur. J. Drug Metab. Pharmacokinet. 1990, 15, 143-53; Balimane and Sinko, Adv. Drug Delivery Rev. 1999, 39, 183-209; Browne, Clin. Neuropharmacol. 1997, 20, 1-12; Bundgaard, Arch. Pharm. Chem. 1979, 86, 1-39; Bundgaard, Controlled Drug Delivery 1987, 17, 179-96; Bundgaard, Adv. Drug Delivery Rev. 1992, 8, 1-38; Fleisher et al., Adv. Drug Delivery Rev. 1996, 19, 115-130; Fleisher et al., Methods Enzymol. 1985, 112, 360-381; Farquhar et al., J. Pharm. Sci. 1983, 72, 324-325; Freeman et al., J. Chem. Soc., Chem. Commun. 1991, 875-877; Friis and Bundgaard, Eur. J. Pharm. Sci. 1996, 4, 49-59; Gangwar et al., Des. Biopharm. Prop. Prodrugs Analogs, 1977, 409-421; Nathwani and Wood, Drugs 1993, 45, 866-94; Sinhababu and Thakker, Adv. Drug Delivery Rev. 1996, 19, 241-273; Stella et al., Drugs 1985, 29, 455-73; Tan et al., Adv. Drug Delivery Rev. 1999, 39, 117-151; Taylor, Adv. Drug Delivery Rev. 1996, 19, 131-148; Valentino and Borchardt, Drug Discovery Today 1997, 2, 148-155; Wiebe and Knaus, Adv. Drug Delivery Rev. 1999, 39, 63-80; and Waller et al., Br. J. Clin. Pharmac. 1989, 28, 497-507.

Methods of Synthesis

The compound provided herein can be prepared, isolated, or obtained by any method known to one of skill in the art, and the following examples are only representative and do not exclude other related procedures.

For example, the compounds of Formula I can be prepared, as shown in Scheme I, via a first aromatic substitution reaction of a trihalo-substituted triazine or pyrimidine with compound I-1 to form compound I-2, which can subsequently be converted to compound I-4 via a second aromatic substitution reaction with compound I-3. Compound I-3 can then be converted to a compound of Formula I via a third aromatic substitution reaction with NH2C(R5aR5bR5c).

Pharmaceutical Compositions

In one embodiment, provided herein is a pharmaceutical composition comprising a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and a pharmaceutically acceptable excipient, adjuvant, carrier, buffer, or stabiliser.

In one embodiment, the pharmaceutical compositions are provided in a dosage form for oral administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers. The pharmaceutical compositions provided herein that are formulated for oral administration may be in tablet, capsule, powder, or liquid form. A tablet may comprise a solid carrier or an adjuvant. Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil, or synthetic oil. Physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol, or polyethylene glycol may be included. A capsule may comprise a solid carrier such as gelatin.

In another embodiment, the pharmaceutical compositions are provided in a dosage form for parenteral administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers. Where pharmaceutical compositions may be formulated for intravenous, cutaneous or subcutaneous injection, the active ingredient will be in the form of a parenterally acceptable aqueous solution, which is pyrogen-free and has a suitable pH, isotonicity, and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles, such as Sodium Chloride injection, Ringer's injection, or Lactated Ringer's injection. Preservatives, stabilisers, buffers, antioxidants, and/or other additives may be included as required.

In yet another embodiment, the pharmaceutical compositions are provided in a dosage form for topical administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers.

The pharmaceutical compositions can also be formulated as modified release dosage forms, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated-, fast-, targeted-, and programmed-release, and gastric retention dosage forms. These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modified-Release Drug Delivery Technology, 2nd Edition, Rathbone et al., Eds., Marcel Dekker, Inc.: New York, N.Y., 2008).

The pharmaceutical compositions provided herein can be provided in a unit-dosage form or multiple-dosage form. A unit-dosage form, as used herein, refers to physically discrete a unit suitable for administration to a human and animal subject, and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of an active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of a unit-dosage form include an ampoule, syringe, and individually packaged tablet and capsule. A unit-dosage form may be administered in fractions or multiples thereof. A multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dosage form. Examples of a multiple-dosage form include a vial, bottle of tablets or capsules, or bottle of pints or gallons.

The pharmaceutical compositions provided herein can be administered at once, or multiple times at intervals of time. It is understood that the precise dosage and duration of treatment may vary with the age, weight, and condition of the patient being treated, and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test or diagnostic data. It is further understood that for any particular individual, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations.

In certain embodiments, the pharmaceutical compositions provided herein further comprise one or more chemotherapeutic agents as defined herein.

A. Oral Administration

The pharmaceutical compositions provided herein for oral administration can be provided in solid, semisolid, or liquid dosage forms for oral administration. As used herein, oral administration also includes buccal, lingual, and sublingual administration. Suitable oral dosage forms include, but are not limited to, tablets, fastmelts, chewable tablets, capsules, pills, strips, troches, lozenges, pastilles, cachets, pellets, medicated chewing gum, bulk powders, effervescent or non-effervescent powders or granules, oral mists, solutions, emulsions, suspensions, wafers, sprinkles, elixirs, and syrups. In addition to the active ingredient(s), the pharmaceutical compositions can contain one or more pharmaceutically acceptable carriers or excipients, including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye-migration inhibitors, sweetening agents, flavoring agents, emulsifying agents, suspending and dispersing agents, preservatives, solvents, non-aqueous liquids, organic acids, and sources of carbon dioxide.

Binders or granulators impart cohesiveness to a tablet to ensure the tablet remaining intact after compression. Suitable binders or granulators include, but are not limited to, starches, such as corn starch, potato starch, and pre-gelatinized starch (e.g., STARCH 1500); gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, panwar gum, ghatti gum, mucilage of isabgol husks, carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone (PVP), Veegum, larch arabogalactan, powdered tragacanth, and guar gum; celluloses, such as ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxyethylcellulose (HEC), hydroxypropylcellulose (HPC), hydroxypropyl methyl cellulose (HPMC); microcrystalline celluloses, such as AVICEL-PH-101, AVICEL-PH-103, AVICEL RC-581, AVICEL-PH-105 (FMC Corp., Marcus Hook, Pa.); and mixtures thereof. Suitable fillers include, but are not limited to, talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof. The amount of a binder or filler in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The binder or filler may be present from about 50 to about 99% by weight in the pharmaceutical compositions provided herein.

Suitable diluents include, but are not limited to, dicalcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar. Certain diluents, such as mannitol, lactose, sorbitol, sucrose, and inositol, when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets. The amount of a diluent in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art.

Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcellulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum and Veegum HV; citrus pulp; cross-linked celluloses, such as croscarmellose; cross-linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystalline cellulose, such as sodium starch glycolate; polacrilin potassium; starches, such as corn starch, potato starch, tapioca starch, and pre-gelatinized starch; clays; aligns; and mixtures thereof. The amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The pharmaceutical compositions provided herein may contain from about 0.5 to about 15% or from about 1 to about 5% by weight of a disintegrant.

Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; mineral oil; light mineral oil; glycerin; sorbitol; mannitol; glycols, such as glycerol behenate and polyethylene glycol (PEG); stearic acid; sodium lauryl sulfate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil; zinc stearate; ethyl oleate; ethyl laureate; agar; starch; lycopodium; silica or silica gels, such as AEROSIL® 200 (W.R. Grace Co., Baltimore, Md.) and CAB-O-SIL® (Cabot Co. of Boston, Mass.); and mixtures thereof. The pharmaceutical compositions provided herein may contain about 0.1 to about 5% by weight of a lubricant.

Suitable glidants include, but are not limited to, colloidal silicon dioxide, CAB-O-SIL® (Cabot Co. of Boston, Mass.), and asbestos-free talc. Suitable coloring agents include, but are not limited to, any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof. A color lake is the combination by adsorption of a water-soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye. Suitable flavoring agents include, but are not limited to, natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation, such as peppermint and methyl salicylate. Suitable sweetening agents include, but are not limited to, sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame. Suitable emulsifying agents include, but are not limited to, gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (TWEEN® 20), polyoxyethylene sorbitan monooleate 80 (TWEEN® 80), and triethanolamine oleate. Suitable suspending and dispersing agents include, but are not limited to, sodium carboxymethylcellulose, pectin, tragacanth, Veegum, acacia, sodium carbomethylcellulose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable preservatives include, but are not limited to, glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol. Suitable wetting agents include, but are not limited to, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether. Suitable solvents include, but are not limited to, glycerin, sorbitol, ethyl alcohol, and syrup. Suitable non-aqueous liquids utilized in emulsions include, but are not limited to, mineral oil and cottonseed oil. Suitable organic acids include, but are not limited to, citric and tartaric acid. Suitable sources of carbon dioxide include, but are not limited to, sodium bicarbonate and sodium carbonate.

It should be understood that many carriers and excipients may serve several functions, even within the same formulation.

The pharmaceutical compositions provided herein for oral administration can be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets. Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach. Enteric-coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation. Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.

The tablet dosage forms can be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.

The pharmaceutical compositions provided herein for oral administration can be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate. The hard gelatin capsule, also known as the dry-filled capsule (DFC), consists of two sections, one slipping over the other, thus completely enclosing the active ingredient. The soft elastic capsule (SEC) is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol. The soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid. The liquid, semisolid, and solid dosage forms provided herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. Capsules containing such solutions can be prepared as described in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545. The capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.

The pharmaceutical compositions provided herein for oral administration can be provided in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups. An emulsion is a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in-water or water-in-oil. Emulsions may include a pharmaceutically acceptable non-aqueous liquid or solvent, emulsifying agent, and preservative. Suspensions may include a pharmaceutically acceptable suspending agent and preservative. Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a di(lower alkyl)acetal of a lower alkyl aldehyde, e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, and hydroalcoholic solutions. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative. For a liquid dosage form, for example, a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.

Other useful liquid and semisolid dosage forms include, but are not limited to, those containing the active ingredient(s) provided herein, and a dialkylated mono- or poly-alkylene glycol, including, 1,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations can further comprise one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.

The pharmaceutical compositions provided herein for oral administration can be also provided in the forms of liposomes, micelles, microspheres, or nanosystems. Micellar dosage forms can be prepared as described in U.S. Pat. No. 6,350,458.

The pharmaceutical compositions provided herein for oral administration can be provided as non-effervescent or effervescent, granules and powders, to be reconstituted into a liquid dosage form. Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders may include diluents, sweeteners, and wetting agents. Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders may include organic acids and a source of carbon dioxide.

Coloring and flavoring agents can be used in all of the above dosage forms.

The pharmaceutical compositions provided herein for oral administration can be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.

B. Parenteral Administration

The pharmaceutical compositions provided herein can be administered parenterally by injection, infusion, or implantation, for local or systemic administration. Parenteral administration, as used herein, include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, intravesical, and subcutaneous administration.

The pharmaceutical compositions provided herein for parenteral administration can be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection. Such dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science (see, Remington: The Science and Practice of Pharmacy, supra).

The pharmaceutical compositions intended for parenteral administration can include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases.

Suitable aqueous vehicles include, but are not limited to, water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection. Suitable non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and medium-chain triglycerides of coconut oil, and palm seed oil. Suitable water-miscible vehicles include, but are not limited to, ethanol, 1,3-butanediol, liquid polyethylene glycol (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerin, N-methyl-2-pyrrolidone, N,N-dimethylacetamide, and dimethyl sulfoxide.

Suitable antimicrobial agents or preservatives include, but are not limited to, phenols, cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoates, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl- and propyl-parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerin, and dextrose. Suitable buffering agents include, but are not limited to, phosphate and citrate. Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable emulsifying agents are those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate. Suitable sequestering or chelating agents include, but are not limited to EDTA. Suitable pH adjusting agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins, including α-cyclodextrin, β-cyclodextrin, hydroxypropyl-β-cyclodextrin, sulfobutylether-β-cyclodextrin, and sulfobutylether 7-β-cyclodextrin (CAPTISOL®, CyDex, Lenexa, Kans.).

When the pharmaceutical compositions provided herein are formulated for multiple dosage administration, the multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as known and practiced in the art.

In one embodiment, the pharmaceutical compositions for parenteral administration are provided as ready-to-use sterile solutions. In another embodiment, the pharmaceutical compositions are provided as sterile dry soluble products, including lyophilized powders and hypodermic tablets, to be reconstituted with a vehicle prior to use. In yet another embodiment, the pharmaceutical compositions are provided as ready-to-use sterile suspensions. In yet another embodiment, the pharmaceutical compositions are provided as sterile dry insoluble products to be reconstituted with a vehicle prior to use. In still another embodiment, the pharmaceutical compositions are provided as ready-to-use sterile emulsions.

The pharmaceutical compositions provided herein for parenteral administration can be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.

The pharmaceutical compositions provided herein for parenteral administration can be formulated as a suspension, solid, semi-solid, or thixotropic liquid, for administration as an implanted depot. In one embodiment, the pharmaceutical compositions provided herein are dispersed in a solid inner matrix, which is surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical compositions diffuse through.

Suitable inner matrixes include, but are not limited to, polymethylmethacrylate, polybutyl-methacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinyl acetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers, such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinyl alcohol, and cross-linked partially hydrolyzed polyvinyl acetate.

Suitable outer polymeric membranes include but are not limited to, polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinyl chloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer.

C. Topical Administration

The pharmaceutical compositions provided herein can be administered topically to the skin, orifices, or mucosa. The topical administration, as used herein, includes (intra)dermal, conjunctival, intracorneal, intraocular, ophthalmic, auricular, transdermal, nasal, vaginal, urethral, respiratory, and rectal administration.

The pharmaceutical compositions provided herein can be formulated in any dosage forms that are suitable for topical administration for local or systemic effect, including emulsions, solutions, suspensions, creams, gels, hydrogels, ointments, dusting powders, dressings, elixirs, lotions, suspensions, tinctures, pastes, foams, films, aerosols, irrigations, sprays, suppositories, bandages, and dermal patches. The topical formulation of the pharmaceutical compositions provided herein can also comprise liposomes, micelles, microspheres, nanosystems, and mixtures thereof.

Pharmaceutically acceptable carriers and excipients suitable for use in the topical formulations provided herein include, but are not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, penetration enhancers, cryoprotectants, lyoprotectants, thickening agents, and inert gases.

The pharmaceutical compositions can also be administered topically by electroporation, iontophoresis, phonophoresis, sonophoresis, or microneedle or needle-free injection, such as POWDERJECT™ (Chiron Corp., Emeryville, Calif.), and BIOJECT™ (Bioject Medical Technologies Inc., Tualatin, Oreg.).

The pharmaceutical compositions provided herein can be provided in the forms of ointments, creams, and gels. Suitable ointment vehicles include oleaginous or hydrocarbon vehicles, including lard, benzoinated lard, olive oil, cottonseed oil, and other oils, white petrolatum; emulsifiable or absorption vehicles, such as hydrophilic petrolatum, hydroxystearin sulfate, and anhydrous lanolin; water-removable vehicles, such as hydrophilic ointment; water-soluble ointment vehicles, including polyethylene glycols of varying molecular weight; emulsion vehicles, either water-in-oil (W/O) emulsions or oil-in-water (O/W) emulsions, including cetyl alcohol, glyceryl monostearate, lanolin, and stearic acid (see, Remington: The Science and Practice of Pharmacy, supra). These vehicles are emollient but generally require addition of antioxidants and preservatives.

Suitable cream base can be oil-in-water or water-in-oil. Suitable cream vehicles may be water-washable, and contain an oil phase, an emulsifier, and an aqueous phase. The oil phase is also called the “internal” phase, which is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol. The aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation may be a nonionic, anionic, cationic, or amphoteric surfactant.

Gels are semisolid, suspension-type systems. Single-phase gels contain organic macromolecules distributed substantially uniformly throughout the liquid carrier. Suitable gelling agents include, but are not limited to, crosslinked acrylic acid polymers, such as carbomers, carboxypolyalkylenes, and CARBOPOL®; hydrophilic polymers, such as polyethylene oxides, polyoxyethylene-polyoxypropylene copolymers, and polyvinylalcohol; cellulosic polymers, such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate, and methylcellulose; gums, such as tragacanth and xanthan gum; sodium alginate; and gelatin. In order to prepare a uniform gel, dispersing agents such as alcohol or glycerin can be added, or the gelling agent can be dispersed by trituration, mechanical mixing, and/or stirring.

The pharmaceutical compositions provided herein can be administered rectally, urethrally, vaginally, or perivaginally in the forms of suppositories, pessaries, bougies, poultices or cataplasm, pastes, powders, dressings, creams, plasters, contraceptives, ointments, solutions, emulsions, suspensions, tampons, gels, foams, sprays, or enemas. These dosage forms can be manufactured using conventional processes as described in Remington: The Science and Practice of Pharmacy, supra.

Rectal, urethral, and vaginal suppositories are solid bodies for insertion into body orifices, which are solid at ordinary temperatures but melt or soften at body temperature to release the active ingredient(s) inside the orifices. Pharmaceutically acceptable carriers utilized in rectal and vaginal suppositories include bases or vehicles, such as stiffening agents, which produce a melting point in the proximity of body temperature, when formulated with the pharmaceutical compositions provided herein; and antioxidants as described herein, including bisulfite and sodium metabisulfite. Suitable vehicles include, but are not limited to, cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol), spermaceti, paraffin, white and yellow wax, and appropriate mixtures of mono-, di- and triglycerides of fatty acids, and hydrogels, such as polyvinyl alcohol, hydroxyethyl methacrylate, and polyacrylic acid. Combinations of the various vehicles can also be used. Rectal and vaginal suppositories may be prepared by compressing or molding. The typical weight of a rectal and vaginal suppository is about 2 to about 3 g.

The pharmaceutical compositions provided herein can be administered ophthalmically in the forms of solutions, suspensions, ointments, emulsions, gel-forming solutions, powders for solutions, gels, ocular inserts, and implants.

The pharmaceutical compositions provided herein can be administered intranasally or by inhalation to the respiratory tract. The pharmaceutical compositions can be provided in the form of an aerosol or solution for delivery using a pressurized container, pump, spray, atomizer, such as an atomizer using electrohydrodynamics to produce a fine mist, or nebulizer, alone or in combination with a suitable propellant, such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane. The pharmaceutical compositions can also be provided as a dry powder for insufflation, alone or in combination with an inert carrier such as lactose or phospholipids; and nasal drops. For intranasal use, the powder can comprise a bioadhesive agent, including chitosan or cyclodextrin.

Solutions or suspensions for use in a pressurized container, pump, spray, atomizer, or nebulizer can be formulated to contain ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active ingredient provided herein; a propellant as solvent; and/or a surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.

The pharmaceutical compositions provided herein can be micronized to a size suitable for delivery by inhalation, such as about 50 micrometers or less, or about 10 micrometers or less. Particles of such sizes can be prepared using a comminuting method known to those skilled in the art, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray drying.

Capsules, blisters, and cartridges for use in an inhaler or insufflator can be formulated to contain a powder mix of the pharmaceutical compositions provided herein; a suitable powder base, such as lactose or starch; and a performance modifier, such as l-leucine, mannitol, or magnesium stearate. The lactose may be anhydrous or in the form of the monohydrate. Other suitable excipients or carriers include, but are not limited to, dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose, and trehalose. The pharmaceutical compositions provided herein for inhaled/intranasal administration can further comprise a suitable flavor, such as menthol and levomenthol; and/or sweeteners, such as saccharin and saccharin sodium.

The pharmaceutical compositions provided herein for topical administration can be formulated to be immediate release or modified release, including delayed-, sustained-, pulsed-, controlled-, targeted, and programmed release.

D. Modified Release

The pharmaceutical compositions provided herein can be formulated as a modified release dosage form. As used herein, the term “modified release” refers to a dosage form in which the rate or place of release of the active ingredient(s) is different from that of an immediate dosage form when administered by the same route. Modified release dosage forms include, but are not limited to, delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms. The pharmaceutical compositions in modified release dosage forms can be prepared using a variety of modified release devices and methods known to those skilled in the art, including, but not limited to, matrix controlled release devices, osmotic controlled release devices, multiparticulate controlled release devices, ion-exchange resins, enteric coatings, multilayered coatings, microspheres, liposomes, and combinations thereof. The release rate of the active ingredient(s) can also be modified by varying the particle sizes and polymorphorism of the active ingredient(s).

Examples of modified release include, but are not limited to, those described in U.S. Pat. Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,639,480; 5,733,566; 5,739,108; 5,891,474; 5,922,356; 5,972,891; 5,980,945; 5,993,855; 6,045,830; 6,087,324; 6,113,943; 6,197,350; 6,248,363; 6,264,970; 6,267,981; 6,376,461; 6,419,961; 6,589,548; 6,613,358; and 6,699,500.

1. Matrix Controlled Release Devices

The pharmaceutical compositions provided herein in a modified release dosage form can be fabricated using a matrix controlled release device known to those skilled in the art (see, Takada et al. in “Encyclopedia of Controlled Drug Delivery,” Vol. 2, Mathiowitz Ed., Wiley, 1999).

In certain embodiments, the pharmaceutical compositions provided herein in a modified release dosage form is formulated using an erodible matrix device, which is water-swellable, erodible, or soluble polymers, including, but not limited to, synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.

Materials useful in forming an erodible matrix include, but are not limited to, chitin, chitosan, dextran, and pullulan; gum agar, gum arabic, gum karaya, locust bean gum, gum tragacanth, carrageenans, gum ghatti, guar gum, xanthan gum, and scleroglucan; starches, such as dextrin and maltodextrin; hydrophilic colloids, such as pectin; phosphatides, such as lecithin; alginates; propylene glycol alginate; gelatin; collagen; cellulosics, such as ethyl cellulose (EC), methylethyl cellulose (MEC), carboxymethyl cellulose (CMC), CMEC, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), cellulose acetate (CA), cellulose propionate (CP), cellulose butyrate (CB), cellulose acetate butyrate (CAB), CAP, CAT, hydroxypropyl methyl cellulose (HPMC), HPMCP, HPMCAS, hydroxypropyl methyl cellulose acetate trimellitate (HPMCAT), and ethyl hydroxyethyl cellulose (EHEC); polyvinyl pyrrolidone; polyvinyl alcohol; polyvinyl acetate; glycerol fatty acid esters; polyacrylamide; polyacrylic acid; copolymers of ethacrylic acid or methacrylic acid (EUDRAGIT®, Rohm America, Inc., Piscataway, N.J.); poly(2-hydroxyethyl-methacrylate); polylactides; copolymers of L-glutamic acid and ethyl-L-glutamate; degradable lactic acid-glycolic acid copolymers; poly-D-(−)-3-hydroxybutyric acid; and other acrylic acid derivatives, such as homopolymers and copolymers of butylmethacrylate, methyl methacrylate, ethyl methacrylate, ethylacrylate, (2-dimethylaminoethyl)methacrylate, and (trimethylaminoethyl)methacrylate chloride.

In certain embodiments, the pharmaceutical compositions provided herein are formulated with a non-erodible matrix device. The active ingredient(s) is dissolved or dispersed in an inert matrix and is released primarily by diffusion through the inert matrix once administered. Materials suitable for use as a non-erodible matrix device include, but are not limited to, insoluble plastics, such as polyethylene, polypropylene, polyisoprene, polyisobutylene, polybutadiene, polymethylmethacrylate, polybutylmethacrylate, chlorinated polyethylene, polyvinylchloride, methyl acrylate-methyl methacrylate copolymers, ethylene-vinyl acetate copolymers, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, vinyl chloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubbers, epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, ethylene/vinyloxyethanol copolymer, polyvinyl chloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, silicone rubbers, polydimethylsiloxanes, and silicone carbonate copolymers; hydrophilic polymers, such as ethyl cellulose, cellulose acetate, crospovidone, and cross-linked partially hydrolyzed polyvinyl acetate; and fatty compounds, such as carnauba wax, microcrystalline wax, and triglycerides.

In a matrix controlled release system, the desired release kinetics can be controlled, for example, via the polymer type employed, the polymer viscosity, the particle sizes of the polymer and/or the active ingredient(s), the ratio of the active ingredient(s) versus the polymer, and other excipients or carriers in the compositions.

The pharmaceutical compositions provided herein in a modified release dosage form can be prepared by methods known to those skilled in the art, including direct compression, dry or wet granulation followed by compression, and melt-granulation followed by compression.

2. Osmotic Controlled Release Devices

The pharmaceutical compositions provided herein in a modified release dosage form can be fabricated using an osmotic controlled release device, including, but not limited to, one-chamber system, two-chamber system, asymmetric membrane technology (AMT), and extruding core system (ECS). In general, such devices have at least two components: (a) a core which contains an active ingredient; and (b) a semipermeable membrane with at least one delivery port, which encapsulates the core. The semipermeable membrane controls the influx of water to the core from an aqueous environment of use so as to cause drug release by extrusion through the delivery port(s).

In addition to the active ingredient(s), the core of the osmotic device optionally includes an osmotic agent, which creates a driving force for transport of water from the environment of use into the core of the device. One class of osmotic agents is water-swellable hydrophilic polymers, which are also referred to as “osmopolymers” and “hydrogels.” Suitable water-swellable hydrophilic polymers as osmotic agents include, but are not limited to, hydrophilic vinyl and acrylic polymers, polysaccharides such as calcium alginate, polyethylene oxide (PEO), polyethylene glycol (PEG), polypropylene glycol (PPG), poly(2-hydroxyethyl methacrylate), poly(acrylic) acid, poly(methacrylic) acid, polyvinylpyrrolidone (PVP), crosslinked PVP, polyvinyl alcohol (PVA), PVA/PVP copolymers, PVA/PVP copolymers with hydrophobic monomers such as methyl methacrylate and vinyl acetate, hydrophilic polyurethanes containing large PEO blocks, sodium croscarmellose, carrageenan, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), carboxymethyl cellulose (CMC) and carboxyethyl, cellulose (CEC), sodium alginate, polycarbophil, gelatin, xanthan gum, and sodium starch glycolate.

The other class of osmotic agents is osmogens, which are capable of imbibing water to affect an osmotic pressure gradient across the barrier of the surrounding coating. Suitable osmogens include, but are not limited to, inorganic salts, such as magnesium sulfate, magnesium chloride, calcium chloride, sodium chloride, lithium chloride, potassium sulfate, potassium phosphates, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, and sodium sulfate; sugars, such as dextrose, fructose, glucose, inositol, lactose, maltose, mannitol, raffinose, sorbitol, sucrose, trehalose, and xylitol; organic acids, such as ascorbic acid, benzoic acid, fumaric acid, citric acid, maleic acid, sebacic acid, sorbic acid, adipic acid, edetic acid, glutamic acid, p-toluenesulfonic acid, succinic acid, and tartaric acid; urea; and mixtures thereof.

Osmotic agents of different dissolution rates can be employed to influence how rapidly the active ingredient(s) is initially delivered from the dosage form. For example, amorphous sugars, such as MANNOGEM™ EZ (SPI Pharma, Lewes, Del.) can be used to provide faster delivery during the first couple of hours to promptly produce the desired therapeutic effect, and gradually and continually release of the remaining amount to maintain the desired level of therapeutic or prophylactic effect over an extended period of time. In this case, the active ingredient(s) is released at such a rate to replace the amount of the active ingredient metabolized and excreted.

The core can also include a wide variety of other excipients and carriers as described herein to enhance the performance of the dosage form or to promote stability or processing.

Materials useful in forming the semipermeable membrane include various grades of acrylics, vinyls, ethers, polyamides, polyesters, and cellulosic derivatives that are water-permeable and water-insoluble at physiologically relevant pHs, or are susceptible to being rendered water-insoluble by chemical alteration, such as crosslinking. Examples of suitable polymers useful in forming the coating, include plasticized, unplasticized, and reinforced cellulose acetate (CA), cellulose diacetate, cellulose triacetate, CA propionate, cellulose nitrate, cellulose acetate butyrate (CAB), CA ethyl carbamate, CAP, CA methyl carbamate, CA succinate, cellulose acetate trimellitate (CAT), CA dimethylaminoacetate, CA ethyl carbonate, CA chloroacetate, CA ethyl oxalate, CA methyl sulfonate, CA butyl sulfonate, CA p-toluene sulfonate, agar acetate, amylose triacetate, beta glucan acetate, beta glucan triacetate, acetaldehyde dimethyl acetate, triacetate of locust bean gum, hydroxylated ethylene-vinylacetate, EC, PEG, PPG, PEG/PPG copolymers, PVP, HEC, HPC, CMC, CMEC, HPMC, HPMCP, HPMCAS, HPMCAT, poly(acrylic) acids and esters and poly-(methacrylic) acids and esters and copolymers thereof, starch, dextran, dextrin, chitosan, collagen, gelatin, polyalkenes, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinyl esters and ethers, natural waxes, and synthetic waxes.

Semipermeable membrane can also be a hydrophobic microporous membrane, wherein the pores are substantially filled with a gas and are not wetted by the aqueous medium but are permeable to water vapor, as disclosed in U.S. Pat. No. 5,798,119. Such hydrophobic but water-vapor permeable membrane are typically composed of hydrophobic polymers such as polyalkenes, polyethylene, polypropylene, polytetrafluoroethylene, polyacrylic acid derivatives, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinylidene fluoride, polyvinyl esters and ethers, natural waxes, and synthetic waxes.

The delivery port(s) on the semipermeable membrane can be formed post-coating by mechanical or laser drilling. Delivery port(s) can also be formed in situ by erosion of a plug of water-soluble material or by rupture of a thinner portion of the membrane over an indentation in the core. In addition, delivery ports can be formed during coating process, as in the case of asymmetric membrane coatings of the type disclosed in U.S. Pat. Nos. 5,612,059 and 5,698,220.

The total amount of the active ingredient(s) released and the release rate can substantially by modulated via the thickness and porosity of the semipermeable membrane, the composition of the core, and the number, size, and position of the delivery ports.

The pharmaceutical compositions in an osmotic controlled-release dosage form can further comprise additional conventional excipients or carriers as described herein to promote performance or processing of the formulation.

The osmotic controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Santus and Baker, J. Controlled Release 1995, 35, 1-21; Verma et al., Drug Development and Industrial Pharmacy 2000, 26, 695-708; Verma et al., J. Controlled Release 2002, 79, 7-27).

In certain embodiments, the pharmaceutical compositions provided herein are formulated as AMT controlled-release dosage form, which comprises an asymmetric osmotic membrane that coats a core comprising the active ingredient(s) and other pharmaceutically acceptable excipients or carriers. See, U.S. Pat. No. 5,612,059 and WO 2002/17918. The AMT controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art, including direct compression, dry granulation, wet granulation, and a dip-coating method.

In certain embodiments, the pharmaceutical compositions provided herein are formulated as ESC controlled-release dosage form, which comprises an osmotic membrane that coats a core comprising the active ingredient(s), a hydroxylethyl cellulose, and other pharmaceutically acceptable excipients or carriers.

3. Multiparticulate Controlled Release Devices

The pharmaceutical compositions provided herein in a modified release dosage form can be fabricated as a multiparticulate controlled release device, which comprises a multiplicity of particles, granules, or pellets, ranging from about 10 μm to about 3 mm, about 50 μm to about 2.5 mm, or from about 100 μm to about 1 mm in diameter. Such multiparticulates can be made by the processes known to those skilled in the art, including wet-and dry-granulation, extrusion/spheronization, roller-compaction, melt-congealing, and by spray-coating seed cores. See, for example, Multiparticulate Oral Drug Delivery; Marcel Dekker: 1994; and Pharmaceutical Pelletization Technology; Marcel Dekker: 1989.

Other excipients or carriers as described herein can be blended with the pharmaceutical compositions to aid in processing and forming the multiparticulates. The resulting particles can themselves constitute the multiparticulate device or can be coated by various film-forming materials, such as enteric polymers, water-swellable, and water-soluble polymers. The multiparticulates can be further processed as a capsule or a tablet.

4. Targeted Delivery

The pharmaceutical compositions provided herein can also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated, including liposome-, resealed erythrocyte-, and antibody-based delivery systems. Examples include, but are not limited to, those disclosed in U.S. Pat. Nos. 6,316,652; 6,274,552; 6,271,359; 6,253,872; 6,139,865; 6,131,570; 6,120,751; 6,071,495; 6,060,082; 6,048,736; 6,039,975; 6,004,534; 5,985,307; 5,972,366; 5,900,252; 5,840,674; 5,759,542; and 5,709,874.

Methods of Use

In one embodiment, provided herein is a method for treating, preventing, or ameliorating one or more symptoms of a PI3K-mediated disorder, disease, or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

In certain embodiments, the PI3K is a wild type PI3K. In certain embodiments, the PI3K is a PI3K variant.

In certain embodiments, the PI3K is a Class I kinase. In certain embodiments, the PI3K is PI3Kα, PI3Kβ, PI3Kδ, or PI3Kγ. In certain embodiments, the PI3K is p111α, p110β, p110δ, or p110γ. In certain embodiments, the PI3K is a wild type of a Class I kinase. In certain embodiments, the PI3K is a variant of a Class I kinase.

In certain embodiments, the PI3K is p110α. In certain embodiments, the PI3K is a wild type of p110α. In certain embodiments, the PI3K is a p110α mutant. In certain embodiments, the p110α mutant is R38H, G106V, K111N, K227E, N345K, C420R, P539R, E542K, E545A, E545G, E545K, Q546K, Q546P, E453Q, H710P, I800L, T1025S, M1043I, M1043V, H1047L, H1047R, or H1047Y. In certain embodiments, the p110α mutant is R38H, K111N, N345K, C420R, P539R, E542K, E545A, E545G, E545K, Q546K, Q546P, I800L, T1025S, M1043I, H1047L, H1047R, or H1047Y. In certain embodiments, the p110α mutant is C420R, E542K, E545A, E545K, Q546K, I800L, M1043I, H1047L, or H1047Y.

In certain embodiments, the PI3K is PI3Kγ. In certain embodiments, the PI3K is a wild type of PI3Kγ. In certain embodiments, the PI3K is a variant of PI3Kγ.

In certain embodiments, the compound provided herein selectively targets PI3Kγ. In certain embodiments, the compound provided herein selectively targets a wild type of PI3Kγ. In certain embodiments, the compound provided herein selectively targets a variant of PI3Kγ.

In certain embodiments, the PI3K is a Class IV kinase. In certain embodiments, the PI3K is a wild type of a Class IV kinase. In certain embodiments, the PI3K is a variant of a Class IV kinase. In certain embodiments, the PI3K is mTOR, ATM, ATR, or DNA-PK. In certain embodiments, the PI3K is mTOR.

In another embodiments, provided herein is a method for treating, preventing, or ameliorating one or more symptoms of a proliferative disease in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

In certain embodiments, the subject is a mammal. In certain embodiments, the subject is a human. In certain embodiments, the subject is a primate other than a human, a farm animal such as cattle, a sport animal, or a pet such as a horse, dog, or cat.

In certain embodiments, the proliferative disease is cancer. In certain embodiments, the proliferative disease is hematological cancer. In certain embodiments, the proliferative disease is an inflammatory disease. In certain embodiments, the proliferative disease is an immune disorder.

The disorders, diseases, or conditions treatable with a compound provided herein, include, but are not limited to, (1) inflammatory or allergic diseases, including systemic anaphylaxis and hypersensitivity disorders, atopic dermatitis, urticaria, drug allergies, insect sting allergies, food allergies (including celiac disease and the like), and mastocytosis; (2) inflammatory bowel diseases, including Crohn's disease, ulcerative colitis, ileitis, and enteritis; (3) vasculitis, and Behcet's syndrome; (4) psoriasis and inflammatory dermatoses, including dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria, viral cutaneous pathologies including those derived from human papillomavirus, HIV or RLV infection, bacterial, flugal, and other parasital cutaneous pathologies, and cutaneous lupus erythematosus; (5) asthma and respiratory allergic diseases, including allergic asthma, exercise induced asthma, allergic rhinitis, otitis media, allergic conjunctivitis, hypersensitivity lung diseases, and chronic obstructive pulmonary disease; (6) autoimmune diseases, including arthritis (including rheumatoid and psoriatic), systemic lupus erythematosus, type I diabetes, myasthenia gravis, multiple sclerosis, Graves' disease, and glomerulonephritis; (7) graft rejection (including allograft rejection and graft-v-host disease), e.g., skin graft rejection, solid organ transplant rejection, bone marrow transplant rejection; (8) fever; (9) cardiovascular disorders, including acute heart failure, hypotension, hypertension, angina pectoris, myocardial infarction, cardiomyopathy, congestive heart failure, atherosclerosis, coronary artery disease, restenosis, and vascular stenosis; (10) cerebrovascular disorders, including traumatic brain injury, stroke, ischemic reperfusion injury and aneurysm; (11) cancers of the breast, skin, prostate, cervix, uterus, ovary, testes, bladder, lung, liver, larynx, oral cavity, colon and gastrointestinal tract (e.g., esophagus, stomach, pancreas), brain, thyroid, blood, and lymphatic system; (12) fibrosis, connective tissue disease, and sarcoidosis, (13) genital and reproductive conditions, including erectile dysfunction; (14) gastrointestinal disorders, including gastritis, ulcers, nausea, pancreatitis, and vomiting; (15) neurologic disorders, including Alzheimer's disease; (16) sleep disorders, including insomnia, narcolepsy, sleep apnea syndrome, and Pickwick Syndrome; (17) pain; (18) renal disorders; (19) ocular disorders, including glaucoma; and (20) infectious diseases, including HIV.

In certain embodiments, the cancer treatable with the methods provided herein includes, but is not limited to, (1) leukemias, including, but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblastic, promyelocytic, myelomonocytic, monocytic, erythroleukemia leukemias and myelodysplastic syndrome or a symptom thereof (such as anemia, thrombocytopenia, neutropenia, bicytopenia or pancytopenia), refractory anemia (RA), RA with ringed sideroblasts (RARS), RA with excess blasts (RAEB), RAEB in transformation (RAEB-T), preleukemia, and chronic myelomonocytic leukemia (CMML), (2) chronic leukemias, including, but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, and hairy cell leukemia; (3) polycythemia vera; (4) lymphomas, including, but not limited to, Hodgkin's disease and non-Hodgkin's disease; (5) multiple myelomas, including, but not limited to, smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary plasmacytoma, and extramedullary plasmacytoma; (6) Waldenstrom's macroglobulinemia; (7) monoclonal gammopathy of undetermined significance; (8) benign monoclonal gammopathy; (9) heavy chain disease; (10) bone and connective tissue sarcomas, including, but not limited to, bone sarcoma, osteosarcoma, chondrosarcoma, Ewing's sarcoma, malignant giant cell tumor, fibrosarcoma of bone, chordoma, periosteal sarcoma, soft-tissue sarcomas, angiosarcoma (hemangiosarcoma), fibrosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, metastatic cancers, neurilemmoma, rhabdomyosarcoma, and synovial sarcoma; (11) brain tumors, including, but not limited to, glioma, astrocytoma, brain stem glioma, ependymoma, oligodendroglioma, nonglial tumor, acoustic neurinoma, craniopharyngioma, medulloblastoma, meningioma, pineocytoma, pineoblastoma, and primary brain lymphoma; (12) breast cancer, including, but not limited to, adenocarcinoma, lobular (small cell) carcinoma, intraductal carcinoma, medullary breast cancer, mucinous breast cancer, tubular breast cancer, papillary breast cancer, primary cancers, Paget's disease, and inflammatory breast cancer; (13) adrenal cancer, including, but not limited to, pheochromocytom and adrenocortical carcinoma; (14) thyroid cancer, including, but not limited to, papillary or follicular thyroid cancer, medullary thyroid cancer, and anaplastic thyroid cancer; (15) pancreatic cancer, including, but not limited to, insulinoma, gastrinoma, glucagonoma, vipoma, somatostatin-secreting tumor, and carcinoid or islet cell tumor; (16) pituitary cancer, including, but limited to, Cushing's disease, prolactin-secreting tumor, acromegaly, and diabetes insipius; (17) eye cancer, including, but not limited, to ocular melanoma such as iris melanoma, choroidal melanoma, and cilliary body melanoma, and retinoblastoma; (18) vaginal cancer, including, but not limited to, squamous cell carcinoma, adenocarcinoma, and melanoma; (19) vulvar cancer, including, but not limited to, squamous cell carcinoma, melanoma, adenocarcinoma, basal cell carcinoma, sarcoma, and Paget's disease; (20) cervical cancers, including, but not limited to, squamous cell carcinoma, and adenocarcinoma; (21) uterine cancer, including, but not limited to, endometrial carcinoma and uterine sarcoma; (22) ovarian cancer, including, but not limited to, ovarian epithelial carcinoma, borderline tumor, germ cell tumor, and stromal tumor; (23) esophageal cancer, including, but not limited to, squamous cancer, adenocarcinoma, adenoid cystic carcinoma, mucoepidermoid carcinoma, adenosquamous carcinoma, sarcoma, melanoma, plasmacytoma, verrucous carcinoma, and oat cell (small cell) carcinoma; (24) stomach cancer, including, but not limited to, adenocarcinoma, fungating (polypoid), ulcerating, superficial spreading, diffusely spreading, malignant lymphoma, liposarcoma, fibrosarcoma, and carcinosarcoma; (25) colon cancer; (26) rectal cancer; (27) liver cancer, including, but not limited to, hepatocellular carcinoma and hepatoblastoma; (28) gallbladder cancer, including, but not limited to, adenocarcinoma; (29) cholangiocarcinomas, including, but not limited to, pappillary, nodular, and diffuse; (30) lung cancer, including, but not limited to, non-small cell lung cancer, squamous cell carcinoma (epidermoid carcinoma), adenocarcinoma, large-cell carcinoma, and small-cell lung cancer; (31) testicular cancer, including, but not limited to, germinal tumor, seminoma, anaplastic, classic (typical), spermatocytic, nonseminoma, embryonal carcinoma, teratoma carcinoma, and choriocarcinoma (yolk-sac tumor); (32) prostate cancer, including, but not limited to, adenocarcinoma, leiomyosarcoma, and rhabdomyosarcoma; (33) penal cancer; (34) oral cancer, including, but not limited to, squamous cell carcinoma; (35) basal cancer; (36) salivary gland cancer, including, but not limited to, adenocarcinoma, mucoepidermoid carcinoma, and adenoidcystic carcinoma; (37) pharynx cancer, including, but not limited to, squamous cell cancer and verrucous; (38) skin cancer, including, but not limited to, basal cell carcinoma, squamous cell carcinoma and melanoma, superficial spreading melanoma, nodular melanoma, lentigo malignant melanoma, and acral lentiginous melanoma; (39) kidney cancer, including, but not limited to, renal cell cancer, adenocarcinoma, hypernephroma, fibrosarcoma, and transitional cell cancer (renal pelvis and/or uterer); (40) Wilms' tumor; (41) bladder cancer, including, but not limited to, transitional cell carcinoma, squamous cell cancer, adenocarcinoma, and carcinosarcoma; and other cancer, including, not limited to, myxosarcoma, osteogenic sarcoma, endotheliosarcoma, lymphangio-endotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma, cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, and papillary adenocarcinomas (See Fishman et al., 1985, Medicine, 2d Ed., J.B. Lippincott Co., Philadelphia and Murphy et al., 1997, Informed Decisions: The Complete Book of Cancer Diagnosis, Treatment, and Recovery, Viking Penguin, Penguin Books U.S.A., Inc., United States of America).

Depending on the disorder, disease, or condition to be treated, and the subject's condition, the compounds or pharmaceutical compositions provided herein can be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracistemal injection or infusion, subcutaneous injection, or implant), inhalation, nasal, vaginal, rectal, sublingual, or topical (e.g., transdermal or local) routes of administration and can be formulated, alone or together, in suitable dosage unit with pharmaceutically acceptable excipients, carriers, adjuvants, and vehicles appropriate for each route of administration. Also provided is administration of the compounds or pharmaceutical compositions provided herein in a depot formulation, in which the active ingredient is released over a predefined time period.

In the treatment, prevention, or amelioration of one or more symptoms of the disorders, diseases, or conditions described herein, an appropriate dosage level generally is ranging from about 0.001 to 100 mg per kg subject body weight per day (mg/kg per day), from about 0.01 to about 75 mg/kg per day, from about 0.1 to about 50 mg/kg per day, from about 0.5 to about 25 mg/kg per day, or from about 1 to about 20 mg/kg per day, which can be administered in single or multiple doses. Within this range, the dosage can be ranging from about 0.005 to about 0.05, from about 0.05 to about 0.5, from about 0.5 to about 5.0, from about 1 to about 15, from about 1 to about 20, or from about 1 to about 50 mg/kg per day.

For oral administration, the pharmaceutical compositions provided herein can be formulated in the form of tablets containing from about 1.0 to about 1,000 mg of the active ingredient, in one embodiment, about 1, about 5, about 10, about 15, about 20, about 25, about 50, about 75, about 100, about 150, about 200, about 250, about 300, about 400, about 500, about 600, about 750, about 800, about 900, and about 1,000 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The pharmaceutical compositions can be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day.

It will be understood, however, that the specific dose level and frequency of dosage for any particular patient can be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.

Also provided herein are methods of modulating PI3K activity, comprising contacting a PIK3 enzyme with a compound provided herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In one embodiment, the PIK3 enzyme is inside a cell.

In certain embodiments, the PI3K is a wild type PI3K. In certain embodiments, the PI3K is a PI3K variant.

In certain embodiments, the PI3K is a Class I kinase. In certain embodiments, the PI3K is PI3Kα, PI3Kβ, PI3Kδ, or PI3Kγ. In certain embodiments, the PI3K is p111α, p110β, p110δ, or p110γ. In certain embodiments, the PI3K is a wild type of a Class I kinase. In certain embodiments, the PI3K is a variant of a Class I kinase.

In certain embodiments, the PI3K is p110α. In certain embodiments, the PI3K is a wild type of p110α. In certain embodiments, the PI3K is a p110α mutant. In certain embodiments, the p110α mutant is R38H, G106V, K111N, K227E, N345K, C420R, P539R, E542K, E545A, E545G, E545K, Q546K, Q546P, E453Q, H710P, I800L, T1025S, M1043I, M1043V, H1047L, H1047R, or H1047Y. In certain embodiments, the p110α mutant is R38H, K111N, N345K, C420R, P539R, E542K, E545A, E545G, E545K, Q546K, Q546P, I800L, T1025S, M1043I, H1047L, H1047R, or H1047Y. In certain embodiments, the p110α mutant is C420R, E542K, E545A, E545K, Q546K, I800L, M1043I, H1047L, or H1047Y.

In certain embodiments, the PI3K is PI3Kγ. In certain embodiments, the PI3K is a wild type of PI3Kγ. In certain embodiments, the PI3K is a variant of PI3Kγ.

In certain embodiments, the compound provided herein selectively targets PI3Kγ. In certain embodiments, the compound provided herein selectively targets a wild type of PI3Kγ. In certain embodiments, the compound provided herein selectively targets a variant of PI3Kγ.

In certain embodiments, the PI3K is a Class IV kinase. In certain embodiments, the PI3K is a wild type of a Class IV kinase. In certain embodiments, the PI3K is a variant of a Class IV kinase. In certain embodiments, the PI3K is mTOR, ATM, ATR, or DNA-PK. In certain embodiments, the PI3K is mTOR.

In certain embodiments, the compound provided herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; shows inhibitory activity against a PI3K and a variant thereof.

In certain embodiments, the compound provided herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; shows inhibitory activity against a wild type of a PI3K. In certain embodiments, the PI3K is PI3Kγ.

In certain embodiments, the compound provided herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; shows inhibitory activity against a PI3K variant. In certain embodiments, the PI3K variant is a p110α mutant. In certain embodiments, the p110α mutant is C420R, E542K, E545A, E545K, Q546K, I800L, M1043I, H1047L, or H1047Y.

The compound provided herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; can also be combined or used in combination with other agents or therapies useful in the treatment, prevention, or amelioration of one or more symptoms of the disorders, diseases, or conditions for which the compounds provided herein are useful, including asthma, allergic rhinitis, eczema, psoriasis, atopic dermatitis, fever, sepsis, systemic lupus erythematosus, diabetes, rheumatoid arthritis, multiple sclerosis, atherosclerosis, transplant rejection, inflammatory bowel disease, cancer, infectious diseases, and those pathologies noted herein.

Suitable other therapeutic agents can also include, but are not limited to, (1) alpha-adrenergic agents; (2) antiarrhythmic agents; (3) anti-atherosclerotic agents, such as ACAT inhibitors; (4) antibiotics, such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; (5) anticancer agents and cytotoxic agents, e.g., alkylating agents, such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; (6) anticoagulants, such as acenocoumarol, argatroban, bivalirudin, lepirudin, fondaparinux, heparin, phenindione, warfarin, and ximelagatran; (7) anti-diabetic agents, such as biguanides (e.g., metformin), glucosidase inhibitors (e.g., acarbose), insulins, meglitinides (e.g., repaglinide), sulfonylureas (e.g., glimepiride, glyburide, and glipizide), thiozolidinediones

(e.g., troglitazone, rosiglitazone, and pioglitazone), and PPAR-gamma agonists; (8) antifungal agents, such as amorolfine, amphotericin B, anidulafungin, bifonazole, butenafine, butoconazole, caspofungin, ciclopirox, clotrimazole, econazole, fenticonazole, filipin, fluconazole, isoconazole, itraconazole, ketoconazole, micafungin, miconazole, naftifine, natamycin, nystatin, oxyconazole, ravuconazole, posaconazole, rimocidin, sertaconazole, sulconazole, terbinafine, terconazole, tioconazole, and voriconazole; (9) antiinflammatories, e.g., non-steroidal anti-inflammatory agents, such as aceclofenac, acemetacin, amoxiprin, aspirin, azapropazone, benorilate, bromfenac, carprofen, celecoxib, choline magnesium salicylate, diclofenac, diflunisal, etodolac, etoricoxib, faislamine, fenbufen, fenoprofen, flurbiprofen, ibuprofen, indometacin, ketoprofen, ketorolac, lomoxicam, loxoprofen, lumiracoxib, meclofenamic acid, mefenamic acid, meloxicam, metamizole, methyl salicylate, magnesium salicylate, nabumetone, naproxen, nimesulide, oxyphenbutazone, parecoxib, phenylbutazone, piroxicam, salicyl salicylate, sulindac, sulfinpyrazone, suprofen, tenoxicam, tiaprofenic acid, and tolmetin; (10) antimetabolites, such as folate antagonists, purine analogues, and pyrimidine analogues; (11) anti-platelet agents, such as GPIIb/IIIa blockers (e.g., abciximab, eptifibatide, and tirofiban), P2Y(AC) antagonists (e.g., clopidogrel, ticlopidine and CS-747), cilostazol, dipyridamole, and aspirin; (12) antiproliferatives, such as methotrexate, FK506 (tacrolimus), and mycophenolate mofetil; (13) anti-TNF antibodies or soluble TNF receptor, such as etanercept, rapamycin, and leflunimide; (14) aP2 inhibitors; (15) beta-adrenergic agents, such as carvedilol and metoprolol; (16) bile acid sequestrants, such as questran; (17) calcium channel blockers, such as amlodipine besylate; (18) chemotherapeutic agents; (19) cyclooxygenase-2 (COX-2) inhibitors, such as celecoxib and rofecoxib; (20) cyclosporins; (21) cytotoxic drugs, such as azathioprine and cyclophosphamide; (22) diuretics, such as chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzothiazide, ethacrynic acid, ticrynafen, chlorthalidone, furosenide, muzolimine, bumetanide, triamterene, amiloride, and spironolactone; (23) endothelin converting enzyme (ECE) inhibitors, such as phosphoramidon; (24) enzymes, such as L-asparaginase; (25) Factor VIIa Inhibitors and Factor Xa Inhibitors; (26) farnesyl-protein transferase inhibitors; (27) fibrates; (28) growth factor inhibitors, such as modulators of PDGF activity; (29) growth hormone secretagogues; (30) HMG CoA reductase inhibitors, such as pravastatin, lovastatin, atorvastatin, simvastatin, NK-104 (a.k.a. itavastatin, nisvastatin, or nisbastatin), and ZD-4522 (also known as rosuvastatin, atavastatin, or visastatin); neutral endopeptidase (NEP) inhibitors; (31) hormonal agents, such as glucocorticoids (e.g., cortisone), estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, and octreotide acetate;

  • (32) immunosuppressants; (33) mineralocorticoid receptor antagonists, such as spironolactone and eplerenone; (34) microtubule-disruptor agents, such as ecteinascidins; (35) microtubule-stabilizing agents, such as pacitaxel, docetaxel, and epothilones A-F; (36) MTP Inhibitors; (37) niacin; (38) phosphodiesterase inhibitors, such as PDE III inhibitors (e.g., cilostazol) and PDE V inhibitors (e.g., sildenafil, tadalafil, and vardenafil); (39) plant-derived products, such as vinca alkaloids, epipodophyllotoxins, and taxanes; (40) platelet activating factor (PAF) antagonists; (41) platinum coordination complexes, such as cisplatin, satraplatin, and carboplatin; (42) potassium channel openers; (43) prenyl-protein transferase inhibitors; (44) protein tyrosine kinase inhibitors; (45) renin inhibitors; (46) squalene synthetase inhibitors; (47) steroids, such as aldosterone, beclometasone, betamethasone, deoxycorticosterone acetate, fludrocortisone, hydrocortisone (cortisol), prednisolone, prednisone, methylprednisolone, dexamethasone, and triamcinolone; (48) TNF-alpha inhibitors, such as tenidap; (49) thrombin inhibitors, such as hirudin; (50) thrombolytic agents, such as anistreplase, reteplase, tenecteplase, tissue plasminogen activator (tPA), recombinant tPA, streptokinase, urokinase, prourokinase, and anisoylated plasminogen streptokinase activator complex (APSAC); (51) thromboxane receptor antagonists, such as ifetroban; (52) topoisomerase inhibitors; (53) vasopeptidase inhibitors (dual NEP-ACE inhibitors), such as omapatrilat and gemopatrilat; and (54) other miscellaneous agents, such as, hydroxyurea, procarbazine, mitotane, hexamethylmelamine, and gold compounds.

In certain embodiments, the other therapies that may be used in combination with the compounds provided herein include, but are not limited to, surgery, endocrine therapy, biologic response modifiers (e.g., interferons, interleukins, and tumor necrosis factor (TNF)), hyperthermia and cryotherapy, and agents to attenuate any adverse effects (e.g., antiemetics).

In certain embodiments, the other therapeutic agents that may be used in combination with the compounds provided herein include, but are not limited to, alkylating drugs (mechlorethamine, chlorambucil, cyclophosphamide, melphalan, and ifosfamide), antimetabolites (cytarabine (also known as cytosine arabinoside or Ara-C), HDAC (high dose cytarabine), and methotrexate), purine antagonists and pyrimidine antagonists (6-mercaptopurine, 5-fluorouracil, cytarbine, and gemcitabine), spindle poisons (vinblastine, vincristine, and vinorelbine), podophyllotoxins (etoposide, irinotecan, and topotecan), antibiotics (daunorubicin, doxorubicin, bleomycin, and mitomycin), nitrosoureas (carmustine and lomustine), enzymes (asparaginase), and hormones (tamoxifen, leuprolide, flutamide, and megestrol), imatinib, adriamycin, dexamethasone, and cyclophosphamide. For a more comprehensive discussion of updated cancer therapies; See, http://www.nci.nih.gov/, a list of the FDA approved oncology drugs at http://www.fda.gov/cder/cancer/druglistframe.htm, and The Merck Manual, Seventeenth Ed. 1999, the entire contents of which are hereby incorporated by reference.

In another embodiment, the method provided herein comprises administration of a compound provided herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof, or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, together with administering one or more chemotherapeutic agents and/or therapies selected from: alkylation agents (e.g., cisplatin, carboplatin); antimetabolites (e.g., methotrexate and 5-FU); antitumour antibiotics (e.g., adriamymycin and bleomycin); antitumour vegetable alkaloids (e.g., taxol and etoposide); antitumor hormones (e.g., dexamethasone and tamoxifen); antitumour immunological agents (e.g., interferon α, β, and γ); radiation therapy; and surgery. In certain embodiments, the one or more chemotherapeutic agents and/or therapies are administered to the subject before, during, or after the administration of the compound provided herein.

Such other agents, or drugs, can be administered, by a route and in an amount commonly used therefor, simultaneously or sequentially with the compound provided herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. When a compound provided herein is used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the compound provided herein can be utilized, but is not required. Accordingly, the pharmaceutical compositions provided herein include those that also contain one or more other active ingredients or therapeutic agents, in addition to a compound provided herein.

The weight ratio of a compound provided herein to the second active ingredient can be varied, and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a compound provided herein is combined with a NSAID, the weight ratio of the compound to the NSAID can range from about 1,000:1 to about 1:1,000, or about 200:1 to about 1:200. Combinations of a compound provided herein and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used.

The compounds provided herein can also be provided as an article of manufacture using packaging materials well known to those of skill in the art. See, e.g., U.S. Pat. Nos. 5,323,907; 5,052,558; and 5,033,252. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.

Provided herein also are kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of active ingredients to a subject. In certain embodiments, the kit provided herein includes a container and a dosage form of a compound provided herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

In certain embodiments, the kit includes a container comprising a dosage form of the compound provided herein, e.g., a compound of Formula I, or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; in a container comprising one or more other therapeutic agent(s) described herein.

Kits provided herein can further include devices that are used to administer the active ingredients. Examples of such devices include, but are not limited to, syringes, needle-less injectors drip bags, patches, and inhalers. The kits provided herein can also include condoms for administration of the active ingredients.

Kits provided herein can further include pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients. For example, if an active ingredient is provided in a solid form that must be reconstituted for parenteral administration, the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration. Examples of pharmaceutically acceptable vehicles include, but are not limited to: aqueous vehicles, including, but not limited to, Water for Injection USP, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles, including, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles, including, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.

The disclosure will be further understood by the following non-limiting examples.

EXAMPLES

As used herein, the symbols and conventions used in these processes, schemes and examples, regardless of whether a particular abbreviation is specifically defined, are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry. Specifically, but without limitation, the following abbreviations may be used in the examples and throughout the specification: g (grams); mg (milligrams); mL (milliliters); L (microliters); M (molar); mM (millimolar); M (micromolar); eq. (equivalent); mmol (millimoles); Hz (Hertz); MHz (megahertz); hr or hrs (hour or hours); min (minutes); and MS (mass spectrometry).

For all of the following examples, standard work-up and purification methods known to those skilled in the art can be utilized. Unless otherwise indicated, all temperatures are expressed in ° C. (degrees Centigrade). All reactions conducted at room temperature unless otherwise noted. Synthetic methodologies illustrated herein are intended to exemplify the applicable chemistry through the use of specific examples and are not indicative of the scope of the disclosure.

Example 1 Synthesis of 4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-6-morpholino-N-(1-phenylcyclopropyl)-1,3,5-triazin-2-amine A1

Compound A1 was prepared according to Scheme 1, where compound 1 (1-[4-chloro-6-(4-morpholinyl)-1,3,5-tianzin-2-yl]-2-(difluoromethyl)-1H-benzimidazole) was synthesized according to the procedure as described in U.S. Pat. Appl. Publ. No. 2007/244110, the disclosure of which is incorporated herein by reference in its entirety.

A mixture of compound 1 (184 mg, 0.502 mmol), 1-phenylcyclopropanamine hydrochloride (170 mg, 1.00 mmol), and potassium carbonate (276 mg, 2.00 mmol) in dioxane (15 mL) was refluxed for 18 hrs. The volatiles were removed in vacuo and the residue was separated by water and ethyl acetate. The organic extracts were dried over anhydrous sodium sulfate and concentrated in vacuo. The crude product was purified by preparative HPLC to give 60 mg (26% yield) of compound A1 as a white solid: 98.5% purity (HPLC); MS m/z: 464.1 (M+1); 1H NMR (CDCl3, 500 MHz) (rotamers) δ 8.43 and 8.13 (2d, J=8.0 and 8.5 Hz, 1H), 7.9 and 7.8 (2d, J=7.5 and 8.0 Hz, 1H), 7.78-7.18 (m, 8H), 6.03 (br s, 1H), 3.91-3.68 (m, 8H), 1.51-1.39 (m, 4H) ppm.

Example 2 Synthesis of 4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-6-morpholino-N-(1-phenylcyclopentyl)-1,3,5-triazin-2-amine A3

Compound A3 was synthesized according to the procedure for compound A1 substituting 1-phenylcyclopentylamine in place of 1-phenylcyclopropanamine hydrochloride. The crude product was purified by preparative HPLC to give 125 mg (51% yield) of compound A3 as a white solid: 98.6% purity (HPLC); MS m/z: 492.3 (M+1); 1H NMR (CDCl3, 500 MHz) (rotamers) δ 8.43 (d, J=8.5 Hz, 0.5H), 7.90 (d, J=8.0 Hz, 0.5H), 7.79 (d, J=8.0 Hz, 0.5H), 7.64 (t, JHF=53.5 Hz, 0.5H), 7.58 (d, J=8.0 Hz, 1H), 7.52-7.38 (m, 3.5H), 7.34-7.25 (m, 2H), 7.22 (t, J=7.5 Hz, 0.5H), 7.15 (t, J=8.0 Hz, 0.5H), 6.92 (t, JHF=53.5 Hz, 0.5H), 5.79 and 5.74 (2s, 1H), 3.88 (m, 2H), 3.81 (m, 2H), 3.75 (m, 1H), 3.68 (m, 1H), 3.49 (m, 2H), 2.45-2.20 (m, 4H), 1.92 (m, 4H) ppm.

Example 3 Synthesis of 4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-6-morpholino-N-(1-phenylcyclohexyl)-1,3,5-triazin-2-amine A4

Compound A4 was synthesized according to the procedure for compound A1 substituting 1-phenylcyclohexylamine in place of 1-phenylcyclopropanamine hydrochloride. The crude product was purified by preparative HPLC to give 184 mg (73% yield) of compound A4: >99.5% purity (HPLC); MS m/z: 506.2 (M+1); 1H NMR (CDCl3, 500 MHz) (rotamers) δ 8.44 (d, J=8.5 Hz, 0.5H), 7.90 (d, J=7.5 Hz, 0.5H), 7.78 (d, J=7.5 Hz, 0.5H), 7.65 (t, JHF=54.0 Hz, 0.5H), 7.53 (d, J=7.5 Hz, 1H), 7.49-7.39 (m, 3.5H), 7.34-7.25 (m, 2H), 7.21 (t, J=7.5 Hz, 0.5H), 7.13 (t, J=7.5 Hz, 0.5H), 6.85 (t, JHF=53.5 Hz, 0.5H), 5.73 and 5.71 (2s, 1H), 3.89 (m, 2H), 3.82 (m, 2H), 3.72 (m, 1H), 3.65 (m, 1H), 3.40 (m, 1H), 3.32 (m, 1H), 2.58-2.36 (m, 2H), 1.95-1.58 (m, 7H), 1.36 (m, 1H) ppm.

Example 4 Synthesis of N-(1-benzylcyclopropyl)-4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-6-morpholino-1,3,5-triazin-2-amine A5

A mixture of 1-benzylcyclopropanamine (73 mg, 0.50 mmol), compound 1 (92 mg, 0.25 mmol) and potassium carbonate (69 mg, 0.50 mmol) in dioxane (10 mL) was refluxed for 2 hrs. The volatiles were removed under vacuum and the residue was purified by prep-HPLC to give compound A5 (41 mg, 34% yield) as a white solid: 99.2% purity (HPLC); MS m/z: 478.2 (M+1); 1H NMR (CDCl3, 500 MHz) (rotamers) δ 8.65 (d, J=8.5 Hz, 0.6H), 8.37 (d, J=7.0 Hz, 0.4H), 7.94 (d, J=8.0 Hz, 0.6H), 7.92 (t, JHF=54.0 Hz, 0.6H), 7.89 (d, J=7.0 Hz, 0.4H), 7.63 (t, JHF=53.5 Hz, 0.4H), 7.51-7.36 (m, 2H), 7.35-7.29 (m, 2H), 7.27-7.22 (m, 1H), 7.22-7.15 (m, 2H), 5.52 and 5.51 (2s, 1H), 4.05-3.69 (m, 8H), 3.06 and 3.00 (2s, 2H), 1.04-0.91 (m, 4H) ppm.

Example 5 Synthesis of N-(1-(4-bromobenzyl)cyclohexyl)-4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-6-morpholino-1,3,5-triazin-2-amine A6

A mixture of compound 1 (100 mg, 0.273 mmol) and 1-(4-bromobenzyl)cyclohexanamine (130 mg, 0.485 mmol) in dioxane (20 mL) was refluxed overnight. The reaction mixture was concentrated under vacuum and the residue was diluted with water and extracted with ethyl acetate. The organic extracts were dried with sodium sulfate and concentrated. The crude product was purified by reverse phase flash chromatography (10% acetonitrile in 0.5% ammonium bicarbonate) to give compound A6 (61 mg, 37% yield) as a white solid: purity: >99.5% purity (HPLC); MS m/z: 598.2 (M+1), 600.2 (M+3); 1H NMR (CDCl3, 500 MHz) (rotamers) δ 8.44 and 8.37 (2d, J=8.0 Hz, 1H), 7.92 and 7.91 (2d, J=7.0 Hz, 1H), 7.65 (t, JHF=54.0 Hz, 1H), 7.50-7.33 (m, 4H), 6.97 and 6.93 (2d, J=8.0 Hz, 1H), 4.95 and 4.85 (2s, 1H), 4.00-3.75 (m, 8H), 3.21 and 3.17 (2s, 2H), 2.19 (m, 2H), 1.78-1.25 (m, 10H) ppm.

Example 6 Synthesis of N-(1-benzylcyclopentyl)-4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-6-morpholino-1,3,5-triazin-2-amine A7

A mixture of 1-benzylcyclopentanol (0.10 g, 0.47 mmol) and 2-chloroacetonitrile (72 mg, 0.95 mmol) in glacial acetic acid (3 mL) was cooled in an ice bath. Concentrated sulfuric acid (0.1 mL) was added and the reaction was stirred at 65° C. overnight. After cooling to room temperature, the reaction mixture was concentrated and the residue was taken up in ethyl acetate. The mixture was washed with water, dried over sodium sulfate, and evaporated to give N-(1-benzylcyclopentyl)-2-chloroacetamide (81 mg, 68% yield) as yellow oil, which was used without further purification: MS m/z: 251 (M+1).

To a mixture of the crude acetamide (80 mg, 0.32 mmol) in dioxane (10 mL) was added hydrochloric acid (6 N, 10 mL) and the mixture was refluxed overnight. After cooling, the reaction mixture was concentrated under vacuum. The pH of the reaction mixture was increased to 10-11 with sodium hydroxide (2M) and was partitioned with ethyl acetate. The combined organic fractions were dried over sodium sulfate and concentrated to give crude 1-benzylcyclopentanamine (42 mg, 74% yield) as a brown oil, which was used directly in the next step. MS m/z: 177 (M+1).

A mixture of the crude amine (42 mg, 0.24 mmol) and compound 1 (100 mg, 0.27 mmol) in dioxane (25 mL) was refluxed overnight. The volatiles were removed under vacuum and the residue was purified by prep-HPLC to give compound A7 (30 mg, 21% yield) as a white solid: 98.6% purity (HPLC); MS m/z: 506.2 (M+1); 1H NMR (CDCl3, 500 MHz) (rotamers) δ 8.54 (d, J=8.5 Hz, 0.2H), 8.36 (d, J=7.0 Hz, 0.8H), 7.92 and 7.89 (2d, J=7.0 Hz, 1H), 7.73 and 7.64 (2t, JHF=54.0 Hz and 53.5 Hz, 1H), 7.49-7.36 (m, 2H), 7.29-7.19 (m, 3H), 7.12-7.03 (m, 2H), 5.12 and 5.08 (2s, 1H), 4.00-3.73 (m, 8H), 3.32 (s, 0.4H), 3.25 (s, 1.6H), 2.02 (m, 2H), 1.88 (m, 2H), 1.79 (m, 4H) ppm.

Example 7 Synthesis of 4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-N-(1-(2-methylbenzyl)cyclopropyl)-6-morpholino-1,3,5-triazin-2-amine A8

A mixture of compound 1 (184 mg, 0.502 mmol), 1-(2-methylbenzyl)-cyclopropanamine hydrochloride (198 mg, 1.00 mmol) and potassium carbonate (207 mg, 1.50 mmol) in dioxane (10 mL) was refluxed overnight. The reaction mixture was concentrated under vacuum. The residue was diluted with a solution of 10% sodium hydroxide and extracted with ethyl acetate. The combined organic fractions were washed with water and brine, dried over sodium sulfate, and concentrated. The crude product was purified by reverse phase flash chromatography (0-70% acetonitrile in 0.01% ammonium bicarbonate) to give compound A8 (160 mg, 65% yield) as a white solid: >99.5% purity (HPLC); MS m/z: 492.3 (M+1); 1H NMR (CDCl3, 500 MHz) (rotamers) δ 8.66 (d, J=8.0 Hz, 0.6H), 8.37 (d, J=7.5 Hz, 0.4H), 7.95 (d, J=7.0 Hz, 0.6H), 7.94 (t, JHF=54.0 Hz, 0.6H), 7.89 (d, J=8.0 Hz, 0.4H), 7.63 (t, JHF=53.0 Hz, 0.4H), 7.52-7.38 (m, 2H), 7.26-7.11 (m, 4H), 5.64-5.60 (m, 1H), 4.20-3.70 (m, 8H), 3.14 and 3.13 (2s, 2H), 2.31 and 2.24 (2s, 3H), 1.05-0.83 (m, 4H) ppm.

Example 8 Synthesis of 4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-N-(1-(3-methylbenzyl)cyclopropyl)-6-morpholino-1,3,5-triazin-2-amine A9

Compound A9 was synthesized according to the procedure for compound A8, substituting 1-(3-methylbenzyl)cyclopropanamine hydrochloride in place of 1-(2-methylbenzyl)cyclopropanamine hydrochloride. The product was purified by reversed phase flash chromatography (0 to 70% acetonitrile in 0.01% ammonium bicarbonate) to give compound A9 (152 mg, 62% yield) as a white solid: >99.5% purity (HPLC); MS m/z: 492.3 (M+1); 1H NMR (CDCl3, 500 MHz) (rotamers) δ 8.64 (d, J=8.0 Hz, 0.6H), 8.35 (d, J=7.0 Hz, 0.4H), 7.93 (d, J=8.0 Hz, 0.6H), 7.90 (t, JHF=54.0 Hz, 0.6H), 7.88 (d, J=6.5 Hz, 0.4H), 7.62 (t, JHF=53.5 Hz, 0.4H), 7.50-7.32 (m, 2H), 7.18 (m, 1H), 7.07 (d, J=7.5 Hz, 0.4H), 7.32 (d, J=7.5 Hz, 0.6H), 6.97 (m, 2H), 5.53 (s, 1H), 4.02-3.60 (m, 8H), 3.01 (s, 0.6H), 2.94 (s, 0.4H), 2.32 (s, 0.4H), 2.31 (s, 0.6H), 1.00-0.80 (m, 4H) ppm.

Example 9 Synthesis of N-(1-(2-chlorobenzyl)cyclopropyl)-4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-6-morpholino-1,3,5-triazin-2-amine A10

Ethylmagnesium bromide (3M in ether, 4.7 mL, 14 mmol) was added to a solution of 2-chlorobenzyl cyanide (1.0 g, 6.6 mmol) and titanium(IV) isopropoxide (2.3 mL, 7.7 mmol) in ether (30 mL) at −78° C. and stirred at that temperature for 10 min. The reaction mixture was warmed to room temperature over an hour. Boron trifluoride diethyl etherate (1.8 mL, 14 mmol) was added and the reaction was stirred for another hour. To the resulting mixture was added successively hydrochloric acid (1M, 15 mL), ether (20 mL) and 10% aqueous sodium hydroxide (25 mL). The reaction mixture was partitioned into ether and the combined organic fractions were dried over sodium sulfate and concentrated under vacuum. The crude product was purified by reverse phase flash chromatography (0-50% acetonitrile in 0.01% aq. trifluoroacetic acid) to give 1-(2-chlorobenzyl)cyclopropanamine trifluoroacetate (187 mg) as a white solid: MS m/z: 182.3 (M+1).

Compound A10 was synthesized according to the procedure for A8, substituting 1-(2-chlorobenzyl)cyclopropanamine trifluoroacetate in place of 1-(2-methylbenzyl)cyclopropanamine hydrochloride. The product was purified by reverse phase flash chromatography (0-65% acetonitrile in 0.01% aq. ammonium bicarbonate) to give compound A10 (214 mg, 14% yield for 2 steps) as a white solid: 99.2% purity (HPLC); MS m/z: 512.2 (M+1); 1H NMR (CDCl3, 500 MHz) (rotamers) δ 8.63 (d, J=8.0 Hz, 0.5H), 8.37 (d, J=7.5 Hz, 0.5H), 7.94 (d, J=7.5 Hz, 0.5H), 7.92 (t, JHF=53.5 Hz, 0.5H), 7.89 (d, J=7.5 Hz, 0.5H), 7.63 (t, JHF=53.5 Hz, 0.5H), 7.51-7.32 (m, 3H), 7.25-7.12 (m, 3H), 5.60 and 5.58 (2s, 1H), 4.02-3.65 (m, 8H), 3.27 and 3.24 (2s, 2H), 1.20-0.85 (m, 4H) ppm.

Example 10 Synthesis of 4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-N-(1-benzylcyclobutyl)-6-morpholino-1,3,5-triazin-2-amine All

Lithium diisopropylamide (2.0 M in tetrahydrofuran, 2.2 mL, 4.4 mmol) was added to a mixture of ethyl isobutyrate (562 mg, 4.39 mmol) in tetrahydrofuran (60 mL) at 78° C. and stirred at this temperature for 1 hr. Then benzyl bromide (500 mg, 2.92 mmol) was added dropwise and the reaction mixture was stirred at −78° C. for another 1 hr. The cold bath was removed and the reaction mixture was stirred at room temperature overnight. The reaction was quenched by the addition of water and the product was extracted with ethyl acetate. The combined organic fractions were washed with water, dried over sodium sulfate, and concentrated under vacuum to give ethyl 1-benzylcyclobutanecarboxylate (620 mg) as a yellow oil, which was used for the next step without further purification.

A mixture of the crude ester (600 mg, 2.75 mmol) in ethanol (30 mL) and sodium hydroxide (2N, 10 mL) was heated to reflux overnight. After cooling, the reaction mixture was concentrated under vacuum. The aqueous solution was acidified with hydrochloric acid (2N) to pH 3-4 and then extracted with ethyl acetate. The combined organic fractions were dried over sodium sulfate and evaporated to give 1-benzyl-cyclobutanecarboxylic acid (340 mg, 65% yield) as a brown oil, which was used directly in the next step: MS m/z: 189 (M−1).

To a mixture of the acid (300 mg, 1.58 mmol) in acetone (30 mL) and water (3 mL) at 0° C. was added triethyl amine (0.33 mL), followed by methyl chloroformate (194 mg, 2.05 mmol). The mixture was stirred at 0° C. for 1 hr and then a solution of sodium azide (154 mg, 2.37 mmol) in water (1 mL) was added dropwise. After stirring at room temperature for another 1 hr, the resulting mixture was diluted with water and extracted with ethyl acetate. The combined organic fractions were washed with water, dried over sodium sulfate, and concentrated under vacuum to give 1-benzylcyclobutanecarbonyl azide (160 mg, 47% yield) as a yellow oil, which was used without further purification.

The crude acyl azide (160 mg, 0.74 mmol) was refluxed in toluene (20 mL) overnight. The solvent was removed under vacuum to give 1,3-bis(1-benzylcyclobutyl)urea (122 mg, 95% yield) as a brown oil, which was used directly in the next step: MS m/z: 347 (M−1).

A mixture of the urea (122 mg, 0.35 mmol) and potassium hydroxide (39 mg, 0.70 mmol) in ethylene glycol (5 mL) was refluxed for 2 hrs. After cooling, the reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic fractions were washed with water, dried over sodium sulfate and evaporated to give 1-benzylcyclobutanamine (110 mg, 97% yield) as a brown oil, which was used without further purification: MS m/z: 162 (M+1).

The crude amine (110 mg, 0.68 mmol) and compound 1 (192 mg, 0.52 mmol) were refluxed in dioxane (25 mL) overnight. The solvent was removed under vacuum and the residue was purified by prep-HPLC to give compound II (60 mg, 41% yield) as a white solid: >99.5% purity (HPLC); MS m/z: 492.2 (M+1); 1H NMR (MeODd4, 500 MHz) δ 8.55 (d, J=8.0 Hz, 1H), 7.89 (t, JHF=53.5 Hz, 1H), 7.80 (d, J=7.5 Hz, 1H), 7.45-7.41 (m, 2H), 7.27-7.13 (m, 5H), 3.92 (m, 4H), 3.81 (m, 4H), 3.33 (s, 2H), 2.45-2.28 (m, 4H), 1.96 (m, 2H) ppm.

Example I A Luciferase-Based Luminescence Assay

PI3K catalyzes the conversion of phosphatidylinositol-4,5-bisphosphate (PIP2) and ATP to phosphatidylinositol-3,4,5-trisphosphate (PIP3) and ADP. PI3K enzymatic activity was determined by measuring the amount of ATP consumed following the kinase reaction using a luciferase-based luminescence assay (Kinase GLO®, Promega Corp., Madison, Wis., USA) in a reaction buffer. The reaction buffer contained 50 mM HEPES, pH 7.5, 3 mM MgCl2, 1 mM EGTA, 100 mM NaCl, 0.03% CHAPS, and 2 mM DTT. Compounds for testing were dissolved and serially diluted in 100% DMSO (total of 10 concentrations), and then diluted 1:25 in the reaction buffer. PI3K enzyme solutions were prepared by diluting PI3K alpha (Invitrogen Corp., Carlsbad, Calif., USA) or PI3K delta (Millipore, Billerica, Mass., USA) in the reaction buffer to 4× the final assay concentration. The final concentrations of enzymes were 1.65 nM and 6.86 nM for PI3K alpha and PI3K delta, respectively. A substrate solution was prepared by mixing PIP2 and ATP in reaction buffer at 2× the final assay concentration. The final concentrations were 50 μM and 25 μM for PIP2 and ATP, respectively. To individual wells of white low volume 384-well assay plates were added 2.5 μl each of the compound and kinase mixtures, followed by shaking. The reactions were started by adding 5 μl of substrate mixture per well and shaking. The assay plates were covered and reactions were allowed to proceed for 1 hour (PI3K alpha) or 2 hours (PI3K delta), after which 10 μl of Kinase GLO® reagent was added. The plates were briefly centrifuged and incubated for 10 minutes, after which luminescence was measured using a FlexStation plate reader (Molecular Devices, Sunnyvale, Calif., USA). IC50 values were determined by curve fitting using Graphpad Prism software (Graphpad Software, La Jolla, Calif., USA).

The biological results are summarized in Table 1, wherein A represents a value no greater than 100 nM, B represents a value greater than 100 nM but less than 200 nM, C represents a value no less than 200 nM but no greater than 500 nM, and D represents a value greater than 500 nM; and wherein A′ represents a ratio of greater than 8, B′ represents a ratio of no less than 4 but no greater than 8, C′ represents a ratio of greater than 2 but less than 4, and D′ represents a ratio of no greater than 2.

TABLE 1 Biological Activity IC50 α/δ Compound p110α p110δ p110β p110γ mTOR ratio Ref. 1 B B D′ A1 C A B′ A3 D D A′ A4 D C A′ A5 D A D D D A′ A6 D D D D D B′ A7 D A B C D A′ A8 D B D D D B′ A10 D C D D D A′ A11 D A A C D A′

In Table 1, the α/δ ratio is the ratio of the IC50 value of a compound against PK3Kα over the IC50 value of the same compound against PK3Kδ; and Ref. 1 is N-benzyl-4-(2-(difluoromethyl)-1H-benzo[d]imidazol-1-yl)-6-morpholino-1,3,5-triazin-2-amine.

The examples set forth above are provided to give those of ordinary skill in the art with a complete disclosure and description of how to make and use the claimed embodiments, and are not intended to limit the scope of what is disclosed herein. Modifications that are obvious to persons of skill in the art are intended to be within the scope of the following claims. All publications, patents, and patent applications cited in this specification are incorporated herein by reference as if each such publication, patent or patent application were specifically and individually indicated to be incorporated herein by reference.

Claims

1. A compound of Formula I:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein: X, Y, and Z are each independently N or CRX, with the proviso that at least two of X, Y, and Z are nitrogen atoms; where RX is hydrogen or C1-6 alkyl; R1 and R2 are each independently (a) hydrogen, cyano, halo, or nitro; (b) C1-16 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, —C(NR1a)NR1bR1c, —OR1a, —OC(O)R1a, —OC(O)OR1a, —OC(O)NR1bR1c, —OC(═NR1a)NR1bR1c, —OS(O)R1a, —OS(O)2R1a, —OS(O)NR1bR1c, —OS(O)2NR1bR1c, —NR1bR1c, —NR1aC(O)R1d, —NR1aC(O)OR1d, —NR1aC(O)NR1bR1c, —NR1aC(═NR1d)NR1bR1c, —NR1aS(O)R1d, —NR1aS(O)2R1d, —NR1aS(O)NR1bR1c, —NR1aS(O)2NR1bR1c, —SR1a, —S(O)R1a, —S(O)2R1a, —S(O)NR1bR1c, or —S(O)2NR1bR1c; wherein each R1a, R1b, R1c, and R1d is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) R1b and R1c together with the N atom to which they are attached form heterocyclyl; R3 and R4 are each independently hydrogen or C1-6 alkyl; or R3 and R4 are linked together to form a bond, C1-6 alkylene, C1-6 heteroalkylene, C2-6 alkenylene, or C2-6 heteroalkenylene; R5a and R5b together with the carbon atom to which they are attached form C3-10 cycloalkyl or heterocyclyl; R5c is C6-14 aryl, heteroaryl, C7-15 aralkyl, or heteroaryl-C1-6 alkyl; and R6 is hydrogen, C1-6 alkyl, —S—C1-6 alkyl, —S(O)—C1-6 alkyl, or —SO2—C1-6 alkyl; wherein each alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaryl-alkyl, and heterocyclyl in R1, R2, R3, R4, R6, RX, R1a, R1b, Rc, R1d, R5a, R5b, and R5c is optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q, wherein each substituent Q is independently selected from (a) oxo, cyano, halo, and nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; and (c) —C(O)Ra, —C(O)ORa, —C(O)NRbRc, —C(NRa)NRbRc, —ORa, —OC(O)Ra, —OC(O)ORa, —OC(O)NRbRc, —OC(═NRa)NRbRc, —OS(O)Ra, —OS(O)2Ra, —OS(O)NRbRc, —OS(O)2NRbRc, —NRbRc, —NRaC(O)Rd, —NRaC(O)ORd, —NRaC(O)NRbRc, —NRaC(═NRd)NRbRc, —NRaS(O)Rd, —NRaS(O)2Rd, —NRaS(O)NRbRc, —NRaS(O)2NRbRc, —SRa, —S(O)Ra, —S(O)2Ra, —S(O)NRbRc, and —S(O)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, which is further optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Qa; wherein each Qa is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) —C(O)Re, —C(O)ORe, —C(O)NRfRg, —C(NRe)NRfRg, —ORe, —OC(O)Re, —OC(O)ORe, —OC(O)NRfRg, —OC(═NRe)NRfRg, —OS(O)Re, —OS(O)2Re, —OS(O)NRfRg, —OS(O)2NRfRg, —NRfRg, —NReC(O)Rh, —NReC(O)ORh, —NReC(O)NRfRg, —NReC(═NRh)NRfRg, —NReS(O)Rh, —NReS(O)2Rh, —NReS(O)NRfRg, —NReS(O)2NRfRg, —SRe, —S(O)Re, —S(O)2Re, —S(O)NRfRg, and —S(O)2NRfRg; wherein each Re, Rf, Rg, and Rh is independently (i) hydrogen; (ii) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and Rg together with the N atom to which they are attached form heterocyclyl.

2. (canceled)

3. (canceled)

4. The compound of claim 1, having the structure of Formula V:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein: V is a bond, —(CH2)r—, —O(CH2)r—, —S(CH2)r—, or —N(R8)(CH2)r—; each R8 is independently (a) hydrogen; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more substituents Q; or (c) —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, —C(NR1a)NR1bR1c, —OR1a, —OC(O)R1a, —OC(O)OR1a, —OC(O)NR1bR1c, —OC(═NR1a)NR1bR1c, —OS(O)R1a, —OS(O)2R1a, —OS(O)NR1bR1c, —OS(O)2NR1bR1c, —NR1bR1c, —NR1aC(O)R1d, —NR1aC(O)OR1d, —NR1aC(O)NR1bR1c, —NR1aC(═NR1d)NR1bR1c, —NR1aS(O)R1d, —NR1aS(O)2R1d, —NR1aS(O)NR1bR1c, —NR1aS(O)2NR1bR1c, —S(O)R1a, —S(O)2R1a, —S(O)NR1bR1c, or —S(O)2NR1bR1c; m and r are each an integer of 0, 1, or 2; and n is an integer of 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.

5. (canceled)

6. (canceled)

7. The compound of claim 1, wherein R5c is C6-14 aryl, optionally substituted with one or more substituents Q.

8. The compound of claim 7, wherein R5c is phenyl or naphthyl, each optionally substituted with one or more substituents Q.

9. (canceled)

10. The compound of claim 1, wherein R5c is heteroaryl, optionally substituted with one or more substituents Q.

11. The compound of claim 10, wherein R5c is monocyclic or bicyclic heteroaryl, each optionally substituted with one or more substituents Q.

12. The compound of claim 10, wherein R5c is 5- or 6-membered heteroaryl, optionally substituted with one or more substituents Q.

13. (canceled)

14. The compound of claim 1, wherein R5c is C7-15 aralkyl, optionally substituted with one or more substituents Q.

15. The compound of claim 14, wherein R5c is benzyl, optionally substituted with one or more substituents Q.

16. The compound of claim 14, wherein R5c is benzyl, optionally substituted with one or more substituents, each of which is independently selected from fluoro, chloro, bromo, and methyl.

17. The compound of claim 1, having the structure of Formula III:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein: R7a, R7b, R7c, R7d, and R7e are each independently (a) hydrogen, cyano, halo, or nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more substituents Q; or (c) —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, —C(NR1a)NR1bR1c, —OR1a, —OC(O)R1a, —OC(O)OR1a, —OC(O)NR1bR1c, —OC(═NR1a)NR1bR1c, —OS(O)R1a, —OS(O)2R1a, —OS(O)NR1bR1c, —OS(O)2NR1bR1c, NR1bR1c, —NR1aC(O)R1d, —NR1aC(O)OR1d, —NR1aC(O)NR1bR1c, —NR1aC(═NR1d)NR1bR1c, —NR1aS(O)R1d, —NR1aS(O)2R1d, —NR1aS(O)NR1bR1c, —NR1aS(O)2NR1bR1c, —SR1a, —S(O)R1a, —S(O)2R1a, —S(O)NR1bR1c, or —S(O)2NR1bR1c; or two of R7a, R7b, R7c, R7d, and R7e that are adjacent to each other form C3-10 cycloalkenyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q.

18. (canceled)

19. (canceled)

20. The compound of claim 4, having the structure of Formula VII:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

21. (canceled)

22. (canceled)

23. The compound of claim 1, having the structure of Formula IX:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein: R7a, R7b, R7c, R7d, and R7e are each independently (a) hydrogen, cyano, halo, or nitro; (b) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more substituents Q; or (c) —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, —C(NR1a)NR1bR1c, —OR1a, —OC(O)R1a, —OC(O)OR1a, —OC(O)NR1bR1c, —OC(═NR1a)NR1bR1c, —OS(O)R1a, —OS(O)2R1a, —OS(O)NR1bR1c, —OS(O)2NR1bR1c, —NR1bR1c, —NR1aC(O)R1d, —NR1aC(O)OR1d, —NR1aC(O)NR1bR1c, —NR1aC(═NRd)NR1bR1c, NR1aS(O)R1d, —NR1aS(O)2R1d, —NR1aS(O)NR1bR1c, —NR1aS(O)2NR1bR1c, —SR1a, —S(O)R1a, —S(O)2R1a, —S(O)NR1bR1c, or —S(O)2NR1bR1c; or two of R7a, R7b, R7c, R7d, and R7e that are adjacent to each other form C3-10 cycloalkenyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q; and k is an integer of 1, 2, 3, 4, 5, or 6.

24. (canceled)

25. (canceled)

26. The compound of claim 4, having the structure of Formula XI:

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein k is an integer of 1, 2, 3, 4, 5, or 6.

27. (canceled)

28. (canceled)

29. The compound of claim 23, wherein k is an integer of 1, 2, or 3.

30. (canceled)

31. The compound of claim 17, wherein R7a is hydrogen, halo, or C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q.

32. The compound of claim 31, wherein R7a is hydrogen, fluoro, chloro, bromo, or methyl.

33. The compound of claim 17, wherein R7b is hydrogen, halo, or C1-6 alkyl, wherein the alkyl is optionally substituted with one or more substituents Q.

34. The compound of claim 33, wherein R7b is hydrogen, fluoro, chloro, bromo, or methyl.

35. The compound of claim 17, wherein R7c is hydrogen, halo, or —OR1a.

36. The compound of claim 35, wherein R7c is chloro or bromo.

37. The compound of claim 35, wherein R7c is —O—C1-6 alkyl, optionally substituted with one or more substituents Q.

38. The compound of claim 17, wherein R7d is hydrogen.

39. The compound of claim 17, wherein R7e is hydrogen.

40. The compound of claim 17, wherein two of R7a, R7b, R7c, R7d, and R7e that are adjacent to each other form C3-10 cycloalkenyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q.

41. The compound of claim 40, wherein R7b and R7b together with the carbon atoms to which they are attached from C6-14 aryl, optionally substituted with one or more substituents Q.

42. The compound of claim 4, wherein V is a bond.

43. The compound of claim 4, wherein m is 0, 1, or 2.

44. The compound of claim 42, wherein V is a bond and m is 0 or 2.

45. The compound of claim 4, wherein V is —(CH2)r—.

46. The compound of claim 45, wherein m is 0, 1, or 2.

47. The compound of claim 4, wherein V is —N(R8)(CH2)r.

48. (canceled)

49. The compound of claim 47, wherein V is —N(CH3)(CH2)r—.

50. (canceled)

51. The compound of claim 45, wherein V is —(CH2)2—.

52. The compound of claim 45, wherein V is —(CH2)2— and m is 1.

53. The compound of claim 47, wherein V is —N(CH3)(CH2)2—.

54. The compound of claim 4, wherein n is 0.

55. The compound of claim 1, wherein R1 is hydrogen or methoxy.

56. (canceled)

57. (canceled)

58. (canceled)

59. The compound of claim 1, wherein R2 is hydrogen or amino.

60. (canceled)

61. (canceled)

62. The compound of claim 1, wherein R3 is hydrogen.

63. The compound of claim 1, wherein R4 is hydrogen.

64. (canceled)

65. The compound of claim 1, wherein R6 is methyl, fluoromethyl, difluoromethyl, or trifluoromethyl.

66. (canceled)

67. The compound of claim 1, wherein X is N or CH.

68. (canceled)

69. (canceled)

70. The compound of claim 1, wherein Y is N or CH.

71. (canceled)

72. (canceled)

73. The compound of claim 1, wherein Z is N or CH.

74. (canceled)

75. (canceled)

76. The compound of claim 1, wherein X, Y, and Z are N.

77. (canceled)

78. The compound of claim 1 selected from the group consisting of:

and enantiomers, mixtures of enantiomers, mixtures of two or more diastereomers, and isotopic variants thereof; and pharmaceutically acceptable salts, solvates, hydrates, and prodrugs thereof.

79. A pharmaceutical composition comprising the compound of claim 1, or an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients.

80. The pharmaceutical composition of claim 79, wherein the composition is formulated for single dose administration.

81. The pharmaceutical composition of claim 79, wherein the composition is formulated as oral, parenteral, or intravenous dosage form.

82. The pharmaceutical composition of claim 81, wherein the oral dosage form is a tablet or capsule.

83. The pharmaceutical composition of claim 79, further comprising a second therapeutic agent.

84. A method for the treatment, prevention, or amelioration of one or more symptoms of a PI3K-mediated disorder, disease, or condition in a subject, which comprises administering to the subject the compound of claim 1.

85. (canceled)

86. (canceled)

87. A method for modulating PI3K enzymatic activity, comprising contacting a PI3K enzyme with the compound of claim 1.

88. (canceled)

89. (canceled)

90. (canceled)

91. The method of claim 87, wherein the PI3K is p110γ.

Patent History
Publication number: 20140088102
Type: Application
Filed: Mar 27, 2012
Publication Date: Mar 27, 2014
Applicant: MEI PHARMA, INC. (San Diego, CA)
Inventors: S. David Brown (San Carlos, CA), David J. Matthews (San Francisco, CA)
Application Number: 14/007,626
Classifications
Current U.S. Class: Plural Ring Nitrogens In The Bicyclo Ring System (514/234.5); 1,3,5-triazine Ring (544/113); Enzyme Inactivation By Chemical Treatment (435/184)
International Classification: C07D 403/04 (20060101); A61K 45/06 (20060101); A61K 31/5377 (20060101);